CA2613283A1 - Methods and compositions for expressing a heterologous protease - Google Patents

Methods and compositions for expressing a heterologous protease Download PDF

Info

Publication number
CA2613283A1
CA2613283A1 CA002613283A CA2613283A CA2613283A1 CA 2613283 A1 CA2613283 A1 CA 2613283A1 CA 002613283 A CA002613283 A CA 002613283A CA 2613283 A CA2613283 A CA 2613283A CA 2613283 A1 CA2613283 A1 CA 2613283A1
Authority
CA
Canada
Prior art keywords
protease
cell
virus
cells
pro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002613283A
Other languages
French (fr)
Inventor
Gregory Duke
George Kemble
James Young
Chengjun Mo
Nisha Hazari
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune Vaccines Inc
Original Assignee
Medimmune Vaccines, Inc.
Gregory Duke
George Kemble
James Young
Chengjun Mo
Nisha Hazari
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medimmune Vaccines, Inc., Gregory Duke, George Kemble, James Young, Chengjun Mo, Nisha Hazari filed Critical Medimmune Vaccines, Inc.
Publication of CA2613283A1 publication Critical patent/CA2613283A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1247DNA-directed RNA polymerase (2.7.7.6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination

Abstract

The present invention provides methods and compositions for expressing a protease or pro-protease in cells that do not naturally express the protease or pro-protease. The invention further provides methods of producing viruses, e.g., influenza viruses, in such cells. The invention also provides methods for increasing the titer of influenza viruses grown in cells that express such a heterologous protease or pro-protease. Additionally, the invention provides a protease from Streptomyces griseus useful in the methods and compositions.

Description

DEMANDE OU BREVET VOLUMINEUX

LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter le Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS

THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME:

NOTE POUR LE TOME / VOLUME NOTE:

METHODS AND COMPOSITIONS FOR EXPRESSING A HETEROLOGOUS
PROTEASE

1. Field of the Invention [0001] In one aspect, the present invention provides methods and coinpositions for expressing a protease or pro-protease in cells that do not naturally express the protease or pro-protease. In other aspects, the invention provides methods of producing viruses, e.g., influenza viruses, in such cells. In other aspects, the invention provides methods for increasing the titer of influenza viruses grown in cells that express such a heterologous protease or pro-protease. In still other aspects, the invention provides a heterologous protease from Sts epton2yces griseus useful in the methods and compositions.
2. Background [0002] Influenza pandemics are defined by a dramatic global increase in morbidity and mortality due to influenza illness. Several factors combine to modulate the severity and extent of the pandemic including the low degree of immunity in the population and the efficiency with which the virus can transmit among humans. The latter is generally influenced not only by the virus itself but the density of the population and ease of travel into and out of a region. The virus responsible for the pandemic is generally a recently emerged antigenic variant that the majority of the population have not had prior experience with and, therefore, have little or no immunity to. In addition, efficient human to huinan transinission is a prerequisite for rapid spread and, in the case of zoonotic introduction of animal viruses into human populations, the virus must adapt to replication in humans and be capable of efficient transmission.
[0003] Pandemic influenza spreads very quickly and can have devastating impact. The most severe pandemic of the 20th century, the 1918 pandeinic, killed over 500,000 U.S. citizens and between 20 to 40 million people worldwide. The pandeinic inay produce waves of disease, with peaks of incidence separated by several weeks to inonths. The relatively rapid onset and spread of pandemic influenza presents several problems for responding to a global attack of this magnitude and imposes overwhelming burdens on einergency responders and health care woricers. Rapid identification and response to the emerging pandeinic is clearly a necessary element of the solution; several prograins are currently in place worldwide to monitor emerging influenza viruses including avian influenza viruses that infrequently cause disease in humans. These surveillance data are used in conjunction with predefined pandemic alert levels in order to identify the likelihood of the threat and provide guidance for an effective response.
[0004] Vaccination is the most iinportant public health measure for preventing disease caused by annual epideinics of influenza. The short interval between identification of a potential pandemic and the onset of significantly increased disease levels present significant challenges for producing sufficient vaccine to protect a large segment of the population.
Having vaccine technology and manufacturing infrastructure in place prior to the einergence of the next pandemic will be critical in aineliorating a significant ainount of illness and death.
The short response times needed to produce a "pandemic vaccine" will not allow for prolonged research or process development to be conducted in order to provide an effective response.
[0005] To date, all commercially available influenza vaccines in the United States have been propagated in embryonated hen's eggs. Although influenza virus grows well in hen's eggs, production of vaccine is dependent on the availability of eggs. Supplies of eggs must be organized, and strains for vaccine production selected months in advance of the next flu season, limiting the flexibility of this approach, and often resulting in delays and shortages in production and distribution. Unfortunately, some influenza vaccine strains, such as the prototype A/Fujian/411/02 strain that circulated during the 2003-04 season, do not replicate well in embryonated chicken eggs, and have to be isolated by cell culture in a costly and time consuming procedure.
[0006] Systeins for producing influenza viruses in cell culture have also been developed in recent years (See, e.g., Furminger. Vaccin.e Production, in Nicholson et al.
(eds) Textbook of Influenza pp. 324-3 32; Merten et al. (1996) Production of influenza virus in cell cultuNes for vaccine preparation, in Cohen & Shafferman (eds) Novel Strategies in Design and Production of Vaccines pp. 141-151). Typically, these methods involve the infection of suitable immortalized host cells with a selected strain of virus. While eliminating many of the difficulties related to vaccine production in hen's eggs, not all pathogenic strains of influenza grow well and can be produced according to established tissue culture methods. In addition, many strains with desirable characteristics, e.g., attenuation, temperature sensitivity and cold adaptation, suitable for production of live attenuated vaccines, have not been successfully grown in tissue culture using established methods.
[0007] Thus, there is a need for 1) manufacturing facilities and procedures needed to produce influenza vaccine from cell culture and 2) development of an effective vaccine technology from cell culture to prevent illness caused by seasonal epidemics of influenza. These procedures and technologies can be rapidly applied to the production and distribution of pandemic vaccine in the event of an iinininent pandeinic.
[0008] One of the many obstacles to be overcome in licensing a cell culture based influenza vaccine is the need for proteolytic cleavage of the hemagglutinin (HA) protein for a newly-formed virus to productively infect a new cell. During infection of an animal, the HA protein is cleaved by a trypsin-like serine protease endogenous to the animal. In culture, endopeptidase activity is frequently insufficient to allow robust replication of the influenza virus. Accordingly, proteases such as trypsin are fiequently added to the culture medium following infection of the cells in culture with an influenza virus of interest to increase viral yield. See, e.g., U.S. Patent No. 5,698,433. However, addition of exogenous proteases to the cell culture medium introduces additional components to the culture medium, adding complexity and the need for additional regulatory review. In addition, the addition of exogenous trypin increases the costs associated with malcing vaccine using cell culture methds. Thus, new methods and compositions are needed for growing influenza virus in culture without the need for addition of exogenous proteases. Further, such methods and compositions must overcome the inherent toxicity of expressing active proteases in cells.
These and other unmet needs are provided by the present invention.
[0009] Citation or discussion of a reference herein shall not be construed as an admission that such is prior art to the present invention. In addition, citation of a patent shall not be construed as an admission of its validity.

3. Summary [0010] The present invention provides cells, referred to herein as "cell(s) of the invention,"
comprising a nucleic acid that encodes a protease or pro-protease, such that the cell expresses a higher level of the protease or pro-protease than would be ordinarily expressed in the cell in the absence of the nucleic acid. In certain embodiments, the cell does not normally express the protease or pro-protease. In certain embodiments, the cell expresses the protease or pro-protease at low levels, for example, at levels less than optimal or desired for a particular biological activity, e.g., culture of viruses, e.g., influenza viruses. In certain aspects, the invention provides a cell comprising a nucleic acid encoding a protease or pro-protease, wherein the nucleic acid encoding the protease or pro-protease is stably integrated into the cell's genome. In other aspects, the invention provides a cell comprising a nucleic acid encoding a protease or pro-protease, wherein the nucleic acid encoding the protease or pro-protease is maintained extrachromasomally. In other aspects, the invention provides a cell comprising a nucleic acid encoding a protease or pro-protease, wherein the nucleic acid encoding the protease or pro-protease is transiently expressed in the cell. In certain embodiments, the cell expresses the protease or pro-protease. In certain embodiments, the cell constitutively expresses the protease or pro-protease. In certain embodiments, the cell inducibly expresses the protease or pro-protease. In certain einbodiments, the cell secretes the protease or pro-protease. In certain embodiments, the cell expresses the protease or pro-protease in the cytosol of the cell.
[0011] In certain embodiments, the protease is a serine protease. In certain embodiments, the serine protease is an S 1 family protease. In certain embodiments, the protease is trypsin. In certain embodiments, the serine protease is a bacterial subtilisin. In certain embodiinents, the protease is SPRT from Streptofnyces griseus. In certain embodiments, the protease is a protease listed in Table 1. In certain embodiments, the protease is a pro-protease. In certain embodiments, the pro-protease is trypsinogen. In certain embodiments, the pro-protease is processed into an active protease.
[0012] In certain einbodiments, expression of the protease or pro-protease is under the control of an inducible promoter. In certain embodiments, the inducible promoter is induced by interferon or a downstreain signaling molecule induced by interferon. In certain einbodiments, the inducible promoter is induced by a tetracycline-regulated expression system. In certain embodiments, expression of the protease or pro-protease is under the control of a constitutively active promoter. In certain embodiinents, the nucleic acid encoding the protease or pro-protease comprises sequence encoding a secretion signal that directs secretion of the protease or pro-protease.
[0013] In certain embodiments, the cell expresses between about 0.1 ng and about 50 g of the protease or pro-protease per inl of cell culture. In certain einbodiments, the cell expresses between about I ng and about 50 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses between about 10 ng and about 50 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses between about 100 ng and about 50 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses between about 1 g and about 50 g of the protease or pro-protease per ml of cell culture. In certain einbodiments, the cell expresses between about 0.1 ng and about 5 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses between about 0.1 ng and about 100 ng of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses between about 0.1 ng and about 10 ng of the protease or pro-protease per ml of cell culture.
In certain einbodiments, the cell expresses between about 0.1 ng and about 1 ng of the protease or pro-protease per ml of cell culture. In certain einbodiinents, the cell expresses an amount of protease or pro-protease sufficient to increase the titer of virus grown in a culture of the cells expressing the protease or pro-protease. In certain einbodiments, the molecular weight of the protease or pro-protease is calculated based on the pro-protease form of the enzyine. In certain embodiments, the molecular weight of the protease or pro-protease is calculated based on the mature, active foim of the protease.

[00141 In certain embodiments, the cell expresses at least about 0.1 ng of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 1 ng of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 10 ng of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 100 ng of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 1 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 10 g of the protease or pro-protease per ml of cell culture. In certain einbodiments, the cell expresses at least about 20 g of the protease or pro-protease per ml of cell culture.
In certain embodiments, the cell expresses at least about 30 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 40 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses at least about 50 g of the protease or pro-protease per ml of cell culture.

[0015j In certain embodiments, the cell is a bacterial cell. In certain embodiments, the bacterial cells is an E. coli cell. In certain embodiments, the cell is a inarnmalian cell. In certain einbodiments, the mammalian cell is a canine cell. In certain embodiments, the canine cell is an MDCK cell. In certain embodiments the MDCK cell is non-tumorigenic. In certain embodiments, the mammalian cell is a primate cell. In certain embodiments, the primate cell is an African green monkey or human cell. In certain embodiments, the cell is an avian cell. In certain eznbodiments, the avian cell is a chicken cell.

[00161 In another aspect, the invention provides a method of producing a virus, comprising infecting a cell of the invention with an virus, culturing the cell under conditions that allow replication of the virus, and collecting virus from the cell culture. In a specific einbodiment, the virus is an influenza virus.

[0017] In another aspect, the invention provides a method of producing a virus, comprising transfecting a cell of the invention with nucleic acids comprising a viral genome, culturing the cell under conditions that allow replication of the virus, and collecting virus from the cell culture. In a specific embodiment, the viral genome is an influenza genome and the virus is an influenza virus. In some embodiunents, the influenza virus correspond to an influenza B
virus. In some embodiments, the influenza virus correspond to an influenza A
virus. In certain einbodiments, the viruses include an attenuated influenza virus, a cold adapted influenza virus, a teinperature sensitive influenza virus, or a virus with any combination of these desirable properties. In one embodiment, the influenza virus is an influenza B/Ann Arbor/l/66 strain virus, e.g., a cold adapted, temperature sensitive, attenuated strain of B/Ann Arbor/l/66. In another embodiment, the influenza virus is an influenza A/Ann Arbor/6/60 strain virus, e.g., a cold adapted, temperature sensitive, attenuated strain of A/Ann Arbor/6/60.

[0018] In certain embodiments, the methods include recovering influenza viruses and using the viruses in the preparation of an immunogenic composition, e.g., a vaccine.
In one einbodiment the virus is capable of eliciting an iirunune response upon administration, e.g., intranasal administration, to a subject. In some embodiments, the viruses used to prepare a vaccine are inactivated prior to administration, in other embodiments, live-attenuated viruses are used to prepare a vaccine. In certain embodiments, recombinant and reassortant influenza A and influenza B viruses are produced according to the methods of the invention. In one embodiment, a vaccine is prepared comprising a live, inactivated, or killed virus derived from a virus produced by the methods of the invention. In one embodiment, viruses produced by the methods of the invention are used to replicate other viruses in cell culture or eggs. In one embodiment, a vaccine is provided that comprises iminunogenic polypeptides derived from a virus produced by the methods of the invention.

[0019] In certain einbodiments, the cell expresses a pro-protease, and the metllod further comprises adding an exogenous protease to the culture medium. In certain embodiments, the exogenous protease is added to a maximum concentration of about 0.1 gg/ml. In certain einbodiments, the exogenous protease is trypsin.

[0020] In certain embodiments, the virus is a DNA virus. In certain embodiments, the virus is an RNA virus. In certain embodiments, the virus is a single-stranded DNA
virus. In certain embodiments, the virus is a double-stranded DNA virus. In certain embodiments, the virus is a positive-sense single-stranded RNA virus. In certain einbodiments, the virus is a negative-sense single-stranded RNA virus. In certain embodiments, the virus is a double-stranded RNA virus. In certain einbodiinents, the virus is a reverse-transcribing virus.
[0021] In another aspect, the invention provides a method of replicating an influenza virus, coinprising infecting a cell of the invention with an influenza virus, culturing the cell under conditions that allow replication of the influenza virus, and collecting influenza virus from the cell culture. In certain embodiments, such conditions do not include exogenously added proteases or pro-proteases, e.g., trypsin or trypsinogen.

[00221 In still another aspect, the invention provides a metliod of replicating an influenza virus, comprising transfecting a cell with nucleic acids encoding an influenza genome, culturing the cell under conditions that allow replication of the influenza virus, and collecting influenza virus from the cell culture. In certain embodiments, such conditions do not include exogenously added proteases or pro-proteases, e.g., trypsin or trypsinogen.

[0023] In certain einbodiments, the cell expresses a pro-protease or enzymatically active protease not normally expressed by the cell. In certain embodiments, the cell is a mammalian cell. In certain einbodiments, the cell is an avian cell. In certain embodiments, the cell is a primate cell, canine cell, hamster cell, mouse cell, or rat cell. In certain embodiments, the cell is an MDCK cell. In certain embodiments, the cell is a Vero cell. In certain embodiinents, the cell is a chicken cell.

[0024] In yet another aspect, the invention provides a method of increasing the titer of influenza virus grown in cell culture, comprising culturing the influenza virus in a cell culture, wherein the cells in the cell culture express a protease or pro-protease that i) is heterologous to the cell, and ii) cleaves a hemagglutinin of the influenza virus, thereby increasing the titer of the influenza virus grown in the cell culture relative to the titer obtained by culturing the influenza virus in cells that do not express a heterologous protease or pro-protease.

[0025] In certain embodiments, the cells stably express the heterologous protease. In certain einbodiments, the cells constitutively express the heterologous protease. In certain embodiments, the cells inducibly express the heterologous protease. In certain embodiments, the heterologous protease is trypsin. In certain embodiments, the cells constitutively express the heterologous pro-protease. In certain embodiments, the cells inducibly express the heterologous pro-protease. In certain embodiments, the pro-protease is trypsinogen. In certain embodiments, the protease is SPRT protease frozn Streptornyces griseus. In certain einbodiments, the pro-protease is prepro-SPRT protease from Sti eptonayces griseus.

[0026] One indication of the ability of a cell to support viral replication is the yield of virus obtained from an infected cell culture. Viral yield can be determined by numerous methods known to one skilled in the art. For example, viral yield can be quantified by determining the concentration of virus present in a sainple according to a median tissue culture infectious dose (TCID50) assay that measures infectious virions. The TCID50 values are often reported as the loglo TCID5n/mL. In one embodiment, the cells expressing a heterologous protease support the replication of influenza viruses (e.g., ca/ts strains) to a loglo TCID50/mL of at least 6.0, or at least 6.2, or at least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8. In a specific embodiment, the cells expressing a heterologous protease support the replication of influenza viruses (e.g., ca/ts strains) to commercially reasonable titers (>107 Log TCID50/mL).

[0027] In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by a loglo TCID50/mL of at least about 0.1, or at least about 0.2, or at least about 0.3, or at least about 0.4, or at least about 0.5, or at least about 0.6, or at least about 0.7, or at least about 0.8, or at least about 0.9, or at least about 1.0, or at least about 1.2, or at least about 1.4, or at least about 1.6, or at least about 1.8, or at least about 2.0, or at least about 2.2, or at least about 2.4, or at least about 2.6, or at least about 2.6, or at least about 2.8, or at least about 3.0, or at least about 3.2, or at least about 3.4, or at least about 3.6, or at least about 3.8, or at least about 4.0, or at least a.bout 4.2, or at least about 4.4, or at least about 4.6, or at least about 4.8, or at least about 5.0, relative to the titer of influenza virus produced in a culture of corresponding cells that do not express a heterologous protease and to which no exogenous protease, e.g., trypsin has been added.

[0028] In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 10%, relative to the titer of influenza virus produced in a culture of corresponding cells that do not express a heterologous protease and to which no exogenous protease, e.g., trypsin has been added. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 25%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 50%. In certain embodiinents, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 100%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 500%. In certain einbodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 1000%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 5000%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 10,000%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 30,000%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 50,000%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 70,000%. In certain embodiments, the titer of the influenza virus that is produced in a cell culture of cells expressing a heterologous protease is increased by at least about 100,000%.

[0029] In certain embodiments, the cell culture medium in which a cell of the invention is cultured is serum-free. In certain embodiments, the cell culture medium in which a cell of the invention is cultured contains serum (e.g., fetal calf serum). In certain embodiments, the cell culture medium in which a cell of the invention is cultured contains no exogenously added animal proteins, such media is often referred to "animal protein free" or "APF" media. In certain einbodiments, the cell culture medium in which a cell of the invention is cultured contains exogenous protease (e.g, porcine trypsin).

[0030] In still another aspect, the invention provides a method for producing a heterologous protease or pro-protease in a cell, wherein said cell is capable of supporting influenza replication, comprising culturing a cell comprising a nucleic acid encoding a protease or pro-protease not normally expressed in the cell under conditions that permit expression of said protease or pro-protease, thereby producing the protease or pro-protease in the cell.

[0031] In certain embodiments, the cell expresses a protease. In certain embodiments, the cell stably expresses a protease. In certain embodiments, the cell expresses a pro-protease. In certain einbodiments, the cell stably expresses a pro-protease. In certain embodiments, the cell secretes the protease or pro-protease into the cell culture medium. In certain embodiments, the cell expresses between about 0.1 ng and about 50 g of the protease or pro-protease per ml of cell culture. In certain embodiments, the cell expresses an amount of protease or pro-protease sufficient to increase the titer of virus grown in a culture of the cells expressing the protease or pro-protease. In certain embodiments, expression of the protease or pro-protease is inducible. In certain embodiments, expression of the protease or pro-protease is constitutive.

[0032] In still another aspect, the invention provides a method for inaking a cell that expresses a protease or pro-protease not norinally expressed by the cell, comprising introducing a nucleic acid encoding a protease or pro-protease operably linked to regulatory elements effective to express the protease or pro-protease in the cell (which may or may not be secreted or released from the cell), thereby making a cell that expresses a protease or pro-protease not nonnally expressed by the cell.

[0033] Any suitable technique and/or vector known by one skilled in the art, witllout .20 limitation, can be used to introduce the nucleic acid into the cell. In certain embodiments, the nucleic acid is introduced into the cell as a plasmid, a cosmid, a viral vector, a bacteriophage, a phagemid, a transposon, or an artificial chromosome. In certain embodiments, the nucleic acid is introduced into the cell as a retroviral vector. In certain embodiments, the nucleic acid is stably maintained in the cell. In other einbodiments, the nucleic acid is transiently maintained.

[0034] In certain embodiments, the nucleic acid encoding the protease or pro-protease further comprises a selectable marker. Methods utilizing a selectable marker to select for those cell stably expressing the nucleic acid encoding the protease or pro-protease are known to one skilled in the art. Any selectable marker known to one skilled in the art to be effective in the cell into which the nucleic acid is introduced can be used in such einbodiments. Thus, in certain embodiments, the selectable inarker is an antibiotic resistance gene.
In certain einbodiments, the selectable marker is a gene in an anabolic pathway that complements a deficiency in the cell. For example, the nucleic acid encoding the protease or pro-protease can be introduced into a cell deficient in synthesis of, for example, an amino acid. The nucleic acid can comprise a gene that complements the deficiency of the cell.
By culturing the cell in medium that does not coinprise the ainino acid, the presence of the nucleic acid coinprising the synthesis gene is selected. Any such gene known to one skilled in the art, without limitation, can be used according to the present invention.

[0035] In yet another aspect, the invention provides an isolated nucleic acid that is at least about 90% identical to SEQ ID NO.:1. In certain embodiments, the isolated nucleic acid coinprises or alternatively consists of SEQ ID NO.:1. In certain embodiments, the isolated nucleic acid hybridizes under hybridization conditions to a nucleic acid encoding SEQ ID
NO.:1 (or the complement thereof). In certain embodiments, the hybridization conditions are stringent hybridization conditions. In certain embodiments, the hybridization conditions are highly stringent hybridization conditions.

[0036] In yet another aspect, the invention provides an isolated polypeptide comprising or alternatively consisiting of the amino acid sequence that is SEQ ID NO.:2.

[0037] In still another aspect, the invention provides an isolated nucleic acid encoding a polypeptide comprising or alternatively consisiting of the amino acid sequence that is SEQ
ID NO.:2.

[0038] In yet another aspect, the invention provides an expression vector comprising a nucleic acid of the invention.

[0039] In still another aspect, the invention provides a cell transfected with an expression vector of the invention.

4. Brief Description of the Fieures [0040] Figure 1 presents replication of ca A/Vietnam/1203/2004 (H5N1) in MDCK
cells.
[0041] Figure 2 presents a table showing amounts of luciferase activity observed in cells transfected with different retroviral vectors.

[00421 Figure 3 presents a graphical representation comparing luciferase activity observed in MDCK cells with the concentration of retroviral particles used to infect the MDCK cells.
[0043] Figure 4 presents a table showing luciferase expression in 12 different single MDCK
clones and two different mixtures of MDCK clones.

[0044] Figure 5 presents a table showing a suinmary of MDCK clones obtained froin viral particles produced in two different packaging cell lines and transfected with three different vectors.

[0045] Figures 6A-6C present graphical representations of influenza viral titers obtained by infecting different MDCK clones (panel A control clone, panel B
AinphoTrypsinogen clone, panel C GP2Typsinogen clone) with MDV-A or ca A/NC influenza strains.
Exogenous trypsin added: 0.0 g/ml, open triangles; 0.1 g/ml on day 1 at day 1, closed circles; 1.0 g/ml at days 1-5 open squares.

[0046] Figures 7A-7B present western blots showing expression of 6xHis-labeled trypsinogen in 12 of 16 MDCK clones.

[0047] Figure 8 presents a table showing inducible luciferase expression in 15 different MDCK clones.

[0048] Figure 9 presents the nucleotide sequence (SEQ ID NO: 1) of the sprT
gene, encoding a serine protease from Stfreptomyces griseus.

[0049] Figure 10 presents the amino acid sequence (SEQ ID NO:2) of a serine protease from Streptoinyces griseus encoded by the sprT gene.

[0050] Figure 11 presents the nucleotide sequence (SEQ ID NO:3) encoding trypsinogen.
[0051] Figure 12 presents the Forward and Reverse primers used to clone the sprT gene (SEQ ID NOS: 5 and 5, respectively).

[0052] Figure 13 presents the schematic map of the pT-Rex-DEST30/Luciferase and pT-Rex-DEST30/Trypsin plasmids transfected into R3/7 clones.

5. Detailed Description of the Invention 5.1 Defmitions [0053] Unless defined otherwise, all scientific and technical terms are understood to have the same meaning as commonly used in the art to which they pertain. For the purpose of the present invention the following terms are defined below.

[0054] The terms "nucleic acid," "polynucleotide," "polynucleotide sequence"
and "nucleic acid sequence" refer to single-stranded or double-stranded deoxyribonucleotide or ribonucleotide polymers, or chimeras or analogues thereof. As used herein, the term optionally includes polymers of analogs of naturally occurring nucleotides having the essential nature of natural nucleotides in that they hybridize to single-stranded nucleic acids in a manner similar to naturally occurring nucleotides (e.g., peptide nucleic acids). Unless otherwise indicated, a particular nucleic acid sequence of this invention encoinpasses complementary sequences, in addition to the sequence explicitly indicated.

[0055] The term "gene" is used broadly to refer to any nucleic acid associated with a biological function. Thus, genes include coding sequences and/or the regulatory sequences required for their expression. The term "gene" applies to a specific genomic sequence, as well as to a cDNA or an mRNA encoded by that genomic sequence.

[0056] Genes also include non-expressed nucleic acid segments that, for example, form recognition sequences for other proteins. Non-expressed regulatory sequences include "promoters" and "enhancers," to which regulatory proteins such as transcription factors bind, resulting in transcription of adjacent or nearby sequences. A "tissue specific" promoter or enhancer is one which regulates transcription in a specific tissue type or cell type, or types.
[0057] The term "vector" refers to plasmids, viral vectors, recombinant nucleic acids and cDNA. A vector can also be a naked RNA polynucleotide, a naked DNA
polynucleotide, a polynucleotide composed of both DNA and RNA within the same strand, a poly-lysine-conjugated DNA or RNA, a peptide-conjugated DNA or RNA, a liposome-conjugated DNA, or the like, that are not autonomously replicating. Most commonly, the vectors of the present invention are plasmids.

[0058] An "expression vector" is a vector, such as a plasmid, which is capable of promoting expression, as well as replication of a nucleic acid incorporated therein.
Typically, the nucleic acid to be expressed is "operably linked" to a promoter and/or enhancer, and is subject to transcription regulatory control by the promoter and/or enhancer.

[0059] A "bi-directional expression vector" is typically characterized by two alternative promoters oriented in the opposite direction relative to a nucleic acid situated between the two promoters, such that expression can be initiated in both orientations resulting in, e.g., transcription of both plus (+) or sense strand, and negative (-) or antisense strand RNAs.
Alternatively, the bi-directional expression vector can be an ambisense vector, in which the viral mRNA and viral genomic RNA (as a cRNA) are expressed from the same strand.

[0060] In the context of the invention, the term "isolated" refers to a biological material, such as a nucleic acid or a protein, which is substantially free from components that normally accompany or interact with it in its naturally occurring environment. The isolated material optionally comprises material not found with the material in its natural environment, e.g., a cell. For example, if the material is in its natural environment, such as a cell, the material has been placed at a location in the cell (e.g., genome or genetic element) not native to a material found in that environment. For example, a naturally occurring nucleic acid (e.g., a coding sequence, a promoter, an enhancer, etc.) becomes isolated if it is introduced by non-naturally occurring means to a locus of the genome (e.g., a vector, such as a plasmid or virus vector, or amplicon) not native to that nucleic acid. Such nucleic acids are also referred to as "heterologous" nucleic acids.

[0061] The term "recombinant" indicates that the material (e.g., a nucleic acid or protein) has been artificially or synthetically (non-naturally) altered by human intervention. The alteration can be performed on the material within, or removed from, its natural enviroiunent or state. Specifically, when referring to a virus, e.g., an influenza virus, the virus is recombinant when it is produced by the expression of a recombinant nucleic acid.

[0062] The term "reassortant," when referring to a virus, indicates that the virus includes genetic and/or polypeptide coinponents derived from more than one parental viral strain or source. For example, a 7:1 reassortant includes 7 viral genomic seginents (or gene segments) derived from a first parental virus, and a single coinplementary viral genomic segment, e.g., encoding hemagglutinin or neuraminidase, from a second parental virus. A 6:2 reassortant includes 6 genomic segments, inost commonly the 6 internal genes from a first parental virus, and two complementary segments, e.g., heinagglutinin and neuraminidase, from a different parental virus.

[0063] The term "introduced" when referring to a heterologous or isolated nucleic acid refers to the incorporation of a nucleic acid into a eukaryotic or prokaryotic cell where the nucleic acid can be incorporated into the genome of the cell (e.g., chromosome, plasmid, plastid or mitochondrial DNA), converted into an autonoinous replicon, or transiently expressed (e.g., transfected mRNA). The term includes such methods as "infection,"
"transfection,"
"transforination" and "transduction." In the context of the invention a variety of methods can be employed to introduce nucleic acids into prokaryotic cells, including electroporation, calciuin phosphate precipitation, lipid mediated transfection (lipofection), etc.

[0064] The term "host cell" means a cell which contains a heterologous nucleic acid, such as a vector, and supports the replication and/or expression of the nucleic acid, and optionally production of one or more encoded products including a polypeptide and/or a virus. Host cells can be prokaryotic cells such as E. coli, or eukaryotic cells such as yeast, insect, amphibian, avian or mammalian cells, including human cells. Exemplary host cells in the context of the invention include Vero (African green monkey kidney) cells, Per.C6 cells (human embryonic retinal cells), BHK (baby hamster kidney) cells, primary chick kidney (PCE-) cells, Madin-Darby Canine Kidney (MDCI,'-) cells, Madin-Darby Bovine Kidney (MDBK) cells, 293 cells (e.g., 293T cells), and COS cells (e.g., COS1, COS7 cells). The term host cell encompasses combinations or mixtures of cells including, e.g., mixed cultures of different cell types or cell lines (e.g., Vero and CEK cells). A co-cultivation of electroporated sf vero cells is described for exainple in PCT/USO4/42669 filed December 22, 2004, which is incorporated by reference in their entirety.

[0065] The expression "artificially engineered" is used herein to indicate that the virus, viral nucleic acid or virally encoded product, e.g., a polypeptide, a vaccine, comprises at least one mutation introduced by recombinant methods, e.g., site directed inutagenesis, PCR
mutagenesis, etc. The expression "artificially engineered" when referring to a virus (or viral component or product) coinprising one or more nucleotide mutations and/or ainino acid substitutions indicates that the viral genome or genome segment encoding the virus (or viral coinponent or product) is not derived from naturally occurring sources, such as a naturally occurring or previously existing laboratory strain of virus produced by non-recoinbinant methods (such as progressive passage at 25 C), e.g., a wild type or cold adapted A/Ann Arbor/6/60 or B/Ann Arbor/1/66strain.

[00661 The term "% sequence identity" is used interchangeably herein with the term "% identity" and refers to the level of amino acid sequence identity between two or more peptide sequences or the level of nucleotide sequence identity between two or more nucleotide sequences, when aligned using a sequence alignment program. For exainple, as used herein, 80% identity means the same thing as 80% sequence identity deterinined by a defined algorithm, and means that a given sequence is at least 80% identical to another length of another sequence. Exemplary levels of sequence identity include, but are not limited to, 60, 70, 80, 85, 90, 95, 98% or more sequence identity to a given sequence.

[00671 The term "% sequence homology" is used interchangeably herein with the terin "% homology" and refers to the level of amino acid sequence homology between two or more peptide sequences or the level of nucleotide sequence homology between two or more nucleotide sequences, when aligned using a sequence alignment program. For example, as used herein, 80% homology means the same thing as 80% sequence homology determined by a defined algorithm, and accordingly a homologue of a given sequence has greater than 80%

sequence homology over a length of the given sequence. Exeinplary levels of sequence homology include, but are not limited to, 60, 70, 80, 85, 90, 95, 98% or more sequence homology to a given sequence.

[0068] Exemplary computer programs which can be used to determine identity between two sequences include, but are not liinited to, the suite of BLAST prograins, e.g., BLASTN, BLASTX, and TBLASTX, BLASTP and TBLASTN, publicly available on the Internet at the NCBI website. See also Altschul et al., 1990, J. Mol. Biol. 215:403-10 (with special reference to the published default setting, i.e., parameters w=4, t= 17) and Altschul et al., 1997, Nucleic Acids Res., 25:3389-3402. Sequence searches are typically carried out using the BLASTP program when evaluating a given amino acid sequence relative to amino acid sequences in the GenBank Protein Sequences and other public databases. The BLASTX
program is preferred for searching nucleic acid sequences that have been translated in all reading frames against amino acid sequences in the GenBaiik Protein Sequences and other public databases. Both BLASTP and BLASTX are run using default parameters of an open gap penalty of 11.0, and an extended gap penalty of 1.0, and utilize the BLOSUM-62 matrix.
See' id.

[0069] A preferred aligninent of selected sequences in order to determine "%
identity"
between two or more sequences, is performed using for exainple, the CLUSTAL-W
program in MacVector version 6.5, operated with default parameters, including an open gap penalty of 10.0, an extended gap penalty of 0.1, and a BLOSUM 30 similarity matrix.

[0070] "Hybridizing specifically to" or "specific hybridization" or "selectively hybridize to", refers to the binding, duplexing, or hybridizing of a nucleic acid molecule preferentially to a particular nucleotide sequence under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA.

[0071] The term "stringent conditions" refers to conditions under which a probe will hybridize preferentially to its target subsequence, and to a lesser extent to, or not at all to, other sequences. "Stringent hybridization" and "stringent hybridization wash conditions" in the context of nucleic acid hybridization experiments such as Southern and northern hybridizations are sequence dependent, and are different under different environinental parameters. An extensive guide to the hybridization of nucleic acids can be found in Tijssen, 1993, Labot=atofy Techniques in Biochernistty azid Molecular Biology -Hybridization with Nucleic Acid Probes, part I, chapter 2, "Overview of principles of hybridization and the strategy of nucleic acid probe assays", Elsevier, NY; Sainbrook et al., 2001, Molecular Cloning: A Laboratoiy Manual, Cold Spring Harbor Laboratory, 3'd ed., NY; and Ausubel et al., eds., Current Edition, Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.

[0072] Generally, highly stringent hybridization and wash conditions are selected to be about 5 C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Very stringent conditions are selected to be equal to the Tm for a particular probe.

[0073] One example of stringent hybridization conditions for hybridization of complementary nucleic acids which have more than about 100 complementary residues on a filter in a Southern or northern blot is 50% formalin with 1 mg of heparin at 42 C, with the hybridization being carried out overnight. An example of highly stringent wash conditions is 0.15 M NaCl at 72 C for about 15 minutes. An example of stringent wash conditions is a 0.2X SSC wash at 65 C for 15 minutes. See Sainbrook et al. for a description of SSC buffer.
A high stringency wash)can be preceded by a low stringency wash to reinove background probe signal. An exemplary medium stringency wash for a duplex of, e.g., more than about 100 nucleotides, is lx SSC at 45 C for 15 minutes. An exemplary low stringency wash for a duplex of, e.g., more than about 100 nucleotides, is 4-6x SSC at 40 C for 15 minutes. In general, a signal to noise ratio of 2x (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.

[0074] The term "about," as used herein, unless otherwi=se indicated, refers to a value that is no more than 10% above or below the value being modified by the term. For example, the term "about 5 g/kg" means a range of from 4.5 g/kg to 5.5 g/kg. As another example, "about 1 hour" means a range of from 48 minutes to 72 minutes.

[0075] The term "stably integrated," as used herein in reference to a nucleic acid, refers to a nucleic acid that has recoinbined with a host cell's genomic nucleic acids and thus become a part of a cell's genome. Stably integrated nucleic acids can comprise a selectable marker to ensure that the stably integrated nucleic acids reinain a part of the cells genoine. Stably integrated nucleic acids need not necessarily remain integrated into the genome at a single location; the nucleic acids can integrate at more than one location and can move from location to location within the genome.

5.2 Cells Expressing a Heterologous Protease or Pro-protease [0076] Influenza virus that contains the precursor hemagglutinin molecule (HAO) on its surface is not capable of fusing with a cell and initiating infection. The HAO
inust be proteolytically cleaved, separating the HA1 and HA2 subunits, to achieve its active form.
Virions with mature HA on the surface actively fuse with a host cell and initiate infection.
Several cell types in vivo, including cells in the airway, contain proteases that activate HA;
however, many cell types used in vitro, including MDCK, do not contain active proteases that can efficiently cleave HA. For these cells, exogenous trypsin has been added to the culture at a concentration that does not negatively impact the cell but allows the HA to be cleaved and activated. See, e.g., US Patent Nos. 5,698,433 and 5,756,341.

[0077] Porcine trypsin has been shown to effectively activate HA and is used routinely, by several influenza investigators for this purpose. In the examples described below, recombinant cells expressing, e.g., porcine trypsin or trypsinogen have been evaluated and selected for their ability to support influenza replication in, e.g., MDCK
cells.

[0078] Thus, in certain embodiments, an active protease or pro-protease expressed from a recombinant system or from the cell itself is contemplated in connection with the invention.
Expression of a cloned protease or pro-protease from a recombinant cell line enables the reduction or reznoval of animal derived products as well as provides the protease or pro-protease in situ enabling the most effective cleavage of the HA molecule.
Alternately, a protease or pro-protease encoded in the cell's genome that is not normally expressed by the cell can be expressed by altering the regulation of expression of the protease or pro-protease.
For example, an promoter, e.g., an inducible promoter, that directs transcription and translation of the protease or pro-protease can be introduced by, e.g., homolgous recombination in the region of the cell's genome that regulates expression of the protease or pro-protease. By selecting a promoter (and/or other regulatory sequences) that is/are active the selected cell type, the protease or pro-protease can be expressed in a cell that does not normally express the protease or or pro-protease.

[0079] Any cell known to be useful for culturing influenza known to one skilled in the art without limitation can be used to generate a cell of the invention. For example, suitable host cells for the replication of influenza virus include, e.g., Vero cells, Per.C6 cells, BHK cells, MDCK cells, 293 cells and COS cells, including 293T cells, COS7 cells.
Further, co-cultures including two or more of the above cell lines, e.g., MDCK cells and either 293T or COS cells can be employed at a ratio, e.g., of 1:1, to improve replication efficiency.
In such embodiments, either or both of the co-cultured cells can express the heterologous protease.
5.2.1 Proteases Expressed by Cells [0080] Any protease known to one skilled in the art to be useful in cleaving the HAO
influenza protein can be expressed by a cell according to the present invention.

[0081] Several proteases can be evaluated for their ability to be produced from a recoinbinant system or engineered into a cell, e.g., an MDCK cell, itself. Suitable proteases and pro-proteases include, but are not limited to, trypsin, trypsinogen, and SPRT.
Additional exeinplaiy proteases that can be used are listed in Table 1, below. Further, active fragments of the proteases can also be used in the cells and methods of the present invention.

[0082]_ The skilled artisan can routinely determine whether a particular protease is suitable for use in the cells and methods of the invention. Typically, such assays involve an assessment of cleavage of a viral protein wherein such cleavage is an important step in the virus's life cycle. Cleavage can be assessed directly, e.g, by monitoring production of two smaller proteins from a larger protein, or indirectly, e.g., by monitoring viral titers produced in the presence of the protease. For example, proteases suitable for cells and/or methods for influenza virus culture can be identified by, e.g., assessing cleavage of the HAO protein or by monitoring viral titer in cell culture comprising the protease.

Table 1: Proteases Enzyme Length SwissProt GenBank S 1 family - SA clan Classificatio (AA) Accession No. Accession No.
achelase I protease: giant silkworm 3.4.21.- 213 (P23604) /A
oth, satumid moth ACH1_LONAC
achelase II protease: giant silkworm 3.4.21.- 214 (P23605) /A
oth, satumid moth ACH2LONAC
acrosin: goat 3.4.21.10 60 (P10626) /A
ACROCAPHI

acrosin: human 3.4.21.10 421 (P10323) Y00970 ACROHUMAN
acrosin: mouse 3.4.21.10 436 (P23578) S66245 ACROMOUSE
acrosin : pig 3.4.21.10 415 (P08001) J04950 ACROPIG

acrosin : rabbit 3.4.21.10 431 (P48038) U05204 ACRORABIT
acrosin: rat 3.4.21.10 437 (p29293) X59254 ACRO RAT

ancrod: malayan pit viper 3.4.21.74 234 (P26324) /A
AGKRH
ANCl ancrod: malayan pit viper 3.4.21.74 258 (P47797) L07308 ancrod: cantil, tropical inocassin 3.4.21.74 20 (P33588) /A
ANCRAGKBI
ancrod: southern copperhead 3.4.21.74 231 (P09872) /A
ANCRAGKCO
apolipoprotein(A): human 3.4.21.- 4548 (P08519) X06290 AP OAHUMAN

apolipoprotein(A): rhesus macaque 3.4.21.- 1420 (P14417) J04635 APOAMACMU
atroxobin: barba amaril, fer-de-lance 3.4.21.74 255 (P04971) J02684 BATXBOTAT
coinplement C1R component: human 3.4.21.41 705 (P00736) X04701 complement C 1 S component: human 3.4.21.42 688 (P09871) X06596 roproteinase E (procarboxypeptidaseA /A 253 (P05805) 4/A
complex: bovine CAC3_BOVIN
azurocidin(cathionic antimicrobial 9/A 251 (P20160) M96326 rotein) : human CAP7 HUMAN
azurocidin(cathionic antimicrobial 4/A 219 (P80015) 9/A
rotein CAP37) : pig CAP7_PIG
calcium-dependent serine proteinase : 3 4.21.- 695 (P15156) X16160 golden hamster CASPMESAU
cathepsin G: human 3.4.21.20 255 (P08311) M16117 CATGHUMAN
cathepsin G: mouse 3.4.21.20 261 g3OUSE 2829) M96801 cathepsin G: rat 3.4.21.20 26 (P 17977) /A

CATGRAT
cerastotin: horned desert viper 3.4.21.- 98 (P81038) /A
CERACERCE

cerastobin: sahara sand viper 3.4.21.- 35 (P18692) /A
CERACERVI
cercarial protease: blood fluke 3.4.21.- 264 (P12546) J03946 CERCSCHMA
complement factor B: bovine 3.4.21.47 16 (P81187) /A
CFABBOVIN

coinplement factor B: huinan 3.4.21.47 764 (P00751) X72875 CFABHUMAN
coinplement factor B: mouse 3.4.21.47 761 (P04186) M60646 CFABMOUSE
complement factor B : pig 3.4.21.47 151 (Q03710) M59240 CFAB PIG
complement factor D: human 3.4.21.46 253 (P00746) M84526 CFAD HUMAN

complement factor D: mouse 3.4.21.46 259 (P03953) Ml 1768 CFADMOUSE
complement factor D : pig 3.4.21.46 259 (P51779) U29948 CFADPIG
compleinent factor D: rat 3.4.21.46 263 (P32038) S73894 CFADRAT

complement factor I: human 3.4.21.45 583 (P05156) y00318 CFAIHUMAN
compleinent factor B-like protease: 3.4.21.- 250 (P81475) /A
chicken CFBL_CHICK
caldecrin: rat 3.4.21.- 268 (P55091) S80379 CLCRRAT
complement C2: human 3.4.21.43 752 g6'MAN 068) M15082 complement C2: mouse 3.4.21.43 760 (P21180) M60579 cocoonase: atlantic horseshoe crab 3.4.21.- 14 (P35586) /A
COCOLIMPO
collagenolytic protease 25 KD II/III: 3.4.21.32 20 (P34153) 4/A
crab-beetle COG1CHIOP
collagenolytic protease 28 KD : red 3.4.21.32 20 (P2073 1) 4/A
ing crab COG1_PARCM
collagenolytic protease 35 KD II: crab- 3.4.21.32 20 (P34154) 4/A
beetle COG2CHIOP
collagenolytic protease 36 KD: crab- 3.4.21.32 20 (P34155) 4/A
eetle COG3CHIOP
collagenolytic protease 36 KD A: red 3.4.21.32 20 (P20732) 9/A
ing crab COGA_PARCM
collagenolytic protease36 KD B red 3.4.21.32 20 (P20733) 9/A
ing crab COGB_PARCM
collagenolytic protease36 KD C red 3.4.21.32 20 (P20734) 9/A
ing crab COGCPARCM
collagenase: cattle grub 3.4.21.- 260 (P08897) X74306 COGSH'YPLI

rachyurin: atlantic sand fiddler crab 3.4.21.32 226 (P00771) /A
COGS UCAPU
complement -activating component of 4/A 699 (P48740) D17525 RA-reactive factor : human CRARHUMAN
complement -activating component of 4/A 704 (P98064) D16492 RA-reactive factor : mouse CRARMOUSE
chyinotrypsinl: african malaria 3.4.21.1 259 (Q27289) Z18887 osquito CTR1_ANOGA
chymotrypsin BI: penoied shrimp, 3.4.21.1 271 (Q00871) X66415 european white shrimp CTR1PENVA
chymotrypsin2: african malaria 3,4.21.1 258 (Q17025) Z18888 os uito CTR2 ANOGA

chymotrypsin2: dog 3.4.21.1 263 (P04813) K01173 chymotrypsinBlI: penoied shrimp, 3.4.21.1 271 (P36178) /A
european white shrimp CTR2_PENVA
chymotrypsinII: european hornet 3.4.21.1 218 (P00769) /A

chymotrypsinII : oriental hornet 3.4.21.1 216 (P00768) N/A

chymotrypsinA: bovine 3.4.21.1 245 (P00766) /A
CTRABOVIN
chymotrypsinA: atlantic cod 3.4.21.1 263 (P47796) X78490 CTRAGADMO
chymotrypsinB: bovine 3.4.21.1 245 (P00767) /A
CTRBBOVIN

chymotrypsinB : atlantic cod 3.4.21.1 245 (P80646) /A
CTRBGADMO
chymotrypsinB: human 3.4.21.1 263 (P17538) M24400 CTRBHUMAN
chymotrypsinB: rat 3.4.21.1 263 (P07338) K02298 CTRBRAT
chymotrypsin-like serine proteinase : 3.4.21.- 254 (P35003) X71438 california red abalone CTRL_HALRU
chymotrypsin-like protease CTRL-1: 3.4.21.- 264 (P40313) X71874 uman CTRLHUMAN
chymotrypsin: penoeid shrimp. 3.4.21.1 31 (P35002) /A
CTRPPENMO
duodenase I: bovine 3.4.21.- 226 (P80219) /A

nite allergen der. F 3: house-dust mite 3.4.21.- 259 (P49275) D63858 ite allergen der. F 6: house-dust mite 3.4.21.- 20 (P49276) /A
DEF6DERFA , ite allergen der. P 3: house-dust mite 3.4.21.- 261 (P39675) U11719 ite allergen der. P 6 house-dust mite 3.4.21.- 20 (P49277) /A

serine protease easter: fruit fly 3.4.21.- 392 (P13582) J03154 EASTDROME

elastase 1: bovine 3.4.21.36 266 (Q28153) M80838 elastase 1: huinan 3.4.21.36 68 (P11423) /A

elastase 1: pig 3.4.21.36 266 (P00772) ELl PIG X04036 elastase 1: rat 3.4.21.36 266 (P00773) V01234 ELlRAT

eutrophil elastase 2A : horse 3.4.21.- 85 (P37357) /A

elastase 2A: human 3.4.21.71 269 (08217) M16631 eutrophil elastase 2B : horse 3.4.21.- 73 (P37358) /A

elastase 2B: human 3.4.21.71 269 (P08218) M16653 elastase 2: bovine 3.4.21.71 269 (Q29461) X97635 elastase 2: mouse 3.4.21.71 271 (P05208) X04573 elastase 2: pig 3.4.21.71 269 (P08419) EL2 PIG M16651 elastase 2: rat 3.4.21.71 271 (p00774) V01233 elastase IIIA: human 3.4.21.70 270 (P09093) M18700 elastase IIIB: human 3.4.21.70 270 (P08861) M16630 elastase : atlantic cod 3.4.21.- 20 (P32197) 'A
ELASGADMO

leukocyteelastase: human 3.4.21.37 267 (P08246) r03545 ELNEHUMAN
enteropeptidase: bovine 3.4.21.9 1035 (P98072) U09859 ENTKBOVIN
enteropeptidase: human 3.4.21.9 1019 (P98073) U09860 ENTKHUMAN

enteropeptidase: mouse 3.4.21.9 1069 (P97435) U73378 ENTKMOUSE
enteropeptidase: pig 3.4.21.9 1034 (P98074) D30799 ENTKPIG
arginine esterase: dog 3.4.21.35 260 (P09582) Y00751 ESTA CANFA

coagulation factor X: bovine 3.4.21.6 492 (P00743) X00673 coagulation factor X: chicken 3.4.21.6 475 (P25155) D00844 coagulation factor X: human 3.4.21.6 488 (P00742) K03194 coagulation factor X: rabbit 3.4.21.6 490 (Tl 4IaBIT 9AF003200 coagulation factor XI: human 3.4.21.27 625 (P03951) M13142 coagulation factor XII: bovine 3.4.21.38 593 (P98140) S70164 coagulation factor XII: guinea pig 3.4.21.38 603 (Q04962) X68615 coagulation factor XII: huinan 3.4.21.38 615 (P00748) M31315 coagulation factor VII: bovine 3.4.21.21 407 (P22457) /A

coagulation factor VII: human 3.4.21.21 466 (P08709) M13232 coagulation factor VII: mouse 3.4.21.21 446 (P70375) U66079 coagulation factor VII: rabbit 3.4.21.21 444 (P98139) U77477 coagulation factor IX: bovine 3.4.21.22 416 (P00741) J00007 coagulation factor IX: dog 3.4.21.22 452 (P 19540) M21757 coagulation factor IX : guinea pig 3.4.21.22 285 (P16295) M26237 coagulation factor IX: human 3.4.21.22 461 (P00740) K02402 coagulation factor IX: mouse 3.4.21.22 459 (P 16294) M23109 coagulation factor IX : pig 3.4.21.22 271 (P 16293) FA9 PIG M26235 coagulation factor IX : rabbit 3.4.21.22 275 (P16292) M26234 coagulation factor IX : rat 3.4.21.22 282 (P16296) M26247 coagulation factor IX : sheep 3.4.21.22 274 (P16291) M26233 flavoxobin: habu 3.4.21.- 260 (P05620) D67078 FLVBTRIFL
gilatoxin: beaded lizard 3.4.21.- 245 (P43685) /A
GILXHELHO
granzyme A: human 3.4.21.78 262 (P12544) M18737 GRAAHUMAN
granzyme A: mouse 3.4.21.78 260 (P11032) X14799 GRAAMOUSE

granzyine B: human 3.4.21.79 247 (P10144) M17016 GRAB HUMAN
granzyme B (G,H): mouse 3.4.21.79 247 g7OUSE 0418) X04072 granzyme C: mouse 3.4.21.- 248 (P08882) M22527 GRACMOUSE
granzyme D: mouse 3.4.21.- 248 (P11033) J03255 GRADMOUSE

granzyme E: mouse 3.4.21.- 248 (P08884) M36901 GRAEMOUSE
granzyme F: mouse 3.4.21.- 248 (P08883) M36902 GRAFMOUSE
granzyine G: mouse 3.4.21.- 248 (P13366) M36900 GRAG MOUSE

granzyme H: human 3.4.21.- 246 (P20718) J02907 GRAHHUMAN

granzyme K: human 3.4.21.- 264 (P49863) U35237 GRAKHUMAN
granzyme K: mouse 3.4.21.- 263 (035205) AF011446 GRAKMOUSE
granzyine K: rat 3.4.21.- 258 (P49864) L19694 GRAKRAT

granzyme M: human 3.4.21.- 257 (P51124) L36936 GRAMHUMAN
granzyme M: rat 3.4.21.- 258 (Q03238) L05175 GRAMRAT
granzyme-like protein I: rat 3.4.21.- 248 (Q06605) X66693 GRLlRAT

granzyme-like protein II: rat 3.4.21.- 248 (Q06606) X68657 alistase: gloydius blornhoffii 3.4.21.- 238 (P81176) /A
HAYSAGKHA
serine protease hepsin : human 3.4.21.- 417 (P05981) M18930 HEPSHUMAN

serine protease hepsin : mouse 3.4.21.- 416 (035453) AF030065 HEPSMOUSE
serine protease hepsin : rat 3.4.21.- 416 (Q05511) X70900 HEPSRAT
epatocyte growth factor activator: 3.4.21.- 655 (Q04756) D14012 uman HGFAHUMAN
epatocyte growth factor -like protein: /A 711 (P26927) M74178 uman HGFL_HUMAN
epatocyte growth factor -like protein: /A 716 (P26928) M74180 ouse HGFLMOUSE
epatocyte growth factor (scatter factor /A 728 (P14210) D90334 ) : human HGFHUMAN
epatocyte growth factor (scatter factor /A 728 (Q08048) D10212 ) : mouse HGFMOUSE
epatocyte growth factor (scatter factor /A 728 (P 17945) D90102 ) : rat HGFRAT
ypodermin A: cattle grub 3.4.21.- 256 (P35587) X74303 HYPAHYPLI
ypodermin B: cattle grub 3.4.21.- 256 (P35588) L24915 HYPBHYPLI
plasma kallikrein: human 3.4.21.34 638 (P03952) M13143 KALHUMAN

lasma kallikrein: mouse 3.4.21.34 638 (P26262) M58588 KALMOUSE
lasma kallikrein: rat 3.4.21.34 638 (P14272) M62357 KAL RAT
glandular kallikrein, submandibular : 3.4.21.35 31 (P12322) /A

guinea pig KLK1_CAVPO

glandular kallikreinl : human 3.4.21.35 262 (P06870) M25629 KLKlHUMAN
glandular kallikreinl: crabeating 3.4.21.35 257 (Q07276) L10039 acaque, cynomolgus monkey KLK1MACFA
glandular kallikrein K1: mouse 3.4.21.35 261 (P15947) M13500 KLKlMOUSE
glandular kallikreinlhainadryas 3.4.21.35 258 (Q28773) L43121 babboon KLK1PAPHA
glandular kallikrein, pancreatic 1: rat 3.4.21.35 261 (P00758) J00758 KLKI RAT
giandular kallikrein, prostatic : guinea 3.4.21.35 239 ~LK2 CAVPO /A
g glandular kallikrein 2: huinan 3.4.21.35 261 (P20151) M18156 onin: rat 3.4.21.35 259 (P00759) M11565 glandular kallikrein K3: mouse 3.4.21.35 261 (P00756) X01389 glandular kallikrein 3, submandibular : 3.4.21.35 188 (P15950) M26534 at KLK3RAT
7S nerve growth factor alpha chain: /A 256 (P00757) X01800 ouse KLK4MOUSE
glandular kallikrein K5: mouse 3.4.21.35 261 (P15945) 00500 glandular kallikrein K6: mouse 3.4.21.35 261 (P00755) V00829 glandular kallikrein 7, 3.4.21.35 261 (P36373) 19647 submandibular/renal: rat KLK7RAT
glandular kallikrein K8: mouse 3.4.21.35 261 (P07628) X03994 glandular kallikrein 8, prostatic: rat 3.4.21.35 261 (P36374) M27217 KI,K8RAT
glandular kallikrein K9: mouse 3.4.21.35 261 (P15949) M17962 glandular kallikrein 9, submandibular: 3.4.21.35 259 (P07647) M11566 at KLK9RAT
glandular kallikrein K11: mouse 3.4.21.35 261 (P15946) X13215 KLKAMOUSE
glandular kallikrein 10: rat 3.4.21.35 244 (P36375) S48142 KLKARAT
glandular kallikrein 12, 3.4.21.35 259 (P36376) M19648 submandibular/renal: rat KLKB RAT
glandular kallikrein K13: mouse 3.4.21.35 261 (P36368) M17982 KLKCMOUSE
E gamma-renin, submandibular gland: (P04071) ouse 3.4.21.54 261 ~~ MOUSE J03877 glandular kallikrein K22: mouse 3.4.21.35 259 (P15948) M17979 KLKLMOUSE
glandular kallikrein, renal: african soft 3.4.21.35 263 (P32824) X17352 f-urred rat K-LKRPRANA
glandular kallikrein 1,' 26: mouse 3.4.21.35 261 (P36369) K01831 KI.,KZMOUSE
glandular kallikrein: pig 3.4.21.35 232 (P00752) /A
KLKPIG
chyinotrypsin-like serine proteinase : 3.4.21.- 26 (P34168) /A
uman LCLPHUMAN
limulus clotting factor C: japanese 3.4.21.84 1019 (P28175) D90271 lorseshoe crab LFC_TACTR
chymase: dog 3.4.21.39 249 (P21842) J02904 chymase: human 3.4.21.39 247 (P23946) M69137 chymase: crabeating macaque, (P56435) cynomolgus monkey 3.4.21.39 247 MCT1_MACFA ~000823 ast cell protease 1: mongolian jird 3.4.21.- 246 (P50340) D45173 ast cell protease 1: mouse 3.4.21.- 246 (P11034) S44609 chymasehamadryas babboon 3.4.21.39 247 (P52195) U38521 ast cell protease I: rat 3.4.21.39 260 (P09650) U67915 nast cell proteaselA: sheep 3.4.21.- 245 (P80931) Y14654 ast cell protease 2: mongolian jird 3.4.21.- 247 (P50341) D45174 nast cell protease 2: mouse 3.4.21.- 244 (P 15119) J05177 ast cell protease II: rat 3.4.21.- 247 (P00770) J02712 ast cell protease3 : mouse 3.4.21.- 21 (P21843) /A

nast cell protease III: rat 3.4.21.- 247 (P50339) D38495 ast cell protease 4: mouse 3.4.21.- 246 (P21812) M55617 ast cell protease 5: mouse 3.4.21.- 247 (P21844) X68805 nast cell protease 6: mouse 3.4.21.- 276 (P21845) M57626 ast cell protease 7: mouse 3.4.21.- 273 (Q02844) L00654 ast cell.protease 7: rat 3.4.21.- 273 (P27435) U67910 ast cell protease 8: mouse 3.4.21.- 247 (P43430) X78545 ast cell protease 9: mouse 3.4.21.- 246 (035164) AF007119 nast cell protease-like protein: mouse 3.4.21.- 246 (Q00356) M57401 MCTXMOUSE

serine protease nudel: fruit fly 3.4.21.- 2616 (P98159) U29153 DLDROME
atural killer cell proteasel: rat 3.4.21.- 248 (P18291) M34097 eutrophil proteinase 4: human 3.4.21.- 25 (P I 8078) /A

okimaxobin I hime-habu 3.4.21.- 20 (P20005) /A
OKIITRIOK
rocl.otting enzyme: japanese horseshoe 3.4.21.86 375 (P21902) M58366 crab PCETACTR
lasmin: bovine 3.4.21.7 812 (P06868) X79402 PLMNBOVIN
plasmin : dog 3.4.21.7 333 (P80009) /A
PLMNCANFA
lasmin: western european hedgehog 3.4.21.7 810 (Q29485) U33171 PLMNERIEU
lasmin: horse 3.4.21.7 338 (P80010) fA
PLMNHORSE
lasmin: huinan 3.4.21.7 810 (P00747) X05199 PLMNHUMAN
lasmin: rhesus macaque 3.4.21.7 810 (P12545) J04697 PLMNMACMU
lasmin: mouse 3.4.21.7 812 (P20918) J04766 PLMNMOUSE
lasmin: sea lamprey 3.4.21.7 325 (P33574) /A
PLMNPETMA
lasmin : pig 3.4.21.7 790 (P06867) /A
PLMNPIG

lasmin: rat 3.4.21.7 169 lasmin: sheep 3.4.21.7 343 (P81286) /A
PLMNSHEEP
yeloblastin: human 3.4.21.76 256 (P24158) X56132 rostate specific antigen: human 3.4.21.77 261 (P07288) X14810 PROSHUMAN
rostate specific antigen: rhesus 3,4.21.35 261 (P33619) X73560 acaque PROSMACMU
itamin-K dependent protein C: bovine 3.4.21.69 456 I(POO745) 1-02435 PRTC BOVIN

itamin-K dependent protein C dog 3.4.21.69 157 (Q28278) D43751 PRTCCANFA

itainin-K dependent protein C goat 3.4.21.69 157 (Q28315) D43752 PRTC CAPHI

itamin-K dependent protein C cat 3.4.21.69 157 (Q28412) D43750 PRTCFELCA
itamin-K dependent protein C horse 3.4.21.69 157 (Q28380) D43753 PRTCHORSE
itamin-K dependent protein C: human 3.4.21.69 461 (P04070) M11228 PRTC_HUMAN
itamin-K dependent protein C: rhesus 3.4.21.69 161 (Q28506) D43754 nacaque PRTC_MACMU
itamin-K dependent protein C: mouse 3.4.21.69 461 (P33587) D10445 PRTCMOUSE
vitamin-K dependent protein C: rabbit 3.4.21.69 458 (Q28661) U49933 PRTCRABIT

vitamin-K dependent protein C: rat 3.4.21.69 461 (P31394) X64336 PRTCRAT
rotease serine-like 1: human 3.4.21.- 276 (043240) AF024605 PSLlHUMAN
rostasin: human 3.4.21.- 343 (Q16651) L41351 rotease M: human 3.4.21.- 244 (Q92876) U62801 roteinase RVV-V alpha: russell viper 3.4.21.95 236 (P18964) /A
RVVA DABRU
roteinase RVV-V gamma: russell 3.4.21.95 236 (P18965) 4/A
viper RVVG_DABRU
serine proteaseSP24D: african malaria 3.4.21.- 271 (Q17004) U21917 osquito S24DANOGA
stratum comeum chymotryptic enzyme: 3.4.21.- 253 (P49862) L33404 uman SCCEHUMAN
serine protease S 1 and 2: fruit fly 3.4.21.- 265 (P 17205) M24379 serine protease3 : fruit fly 3.4.21.- 61 (P17207) M24380 serine protease snake: fruit fly 3.4.21.- 430 (P05049) X04513 SNAKDROME

serine proteinase stubble : fruit fly 3.4.21.- 786 (Q05319) Ll 1451 STUB DROME
Subtilisin: Bacillus subtilis 3.4.21.- 378 /A M28537 hrombin: bovine 3.4.21.5 625 (P00735) V00135 THRBBOVIN
hrombin: huinan 3.4.21.5 622 (P00734) M17262 THRBHUMAN
thrombin: mouse 3.4.21.5 618 (X52308 THRB MOUSE

(P 18292) hrombin: rat 3.4.21.5 617 THRBRAT X52835 gyroxin analog: bushmaster 3.4.21.74 228 (P33589) /A
THRLLACMU
transmembrane protease, serine2 : 3.4.21.- 492 (015393) U75329 uman TMS2HUMAN
trypsinl: african inalaria inosquito 3.4.21.4 274 (P35035) Z18889 TRYI ANOGA
rypsin,cationic: bovine 3.4.21.4 243 (P00760) D38507 rypsin,cationic: dog 3.4.21.4 246 (P06871) M11590 TRYlCANFA
rypsin I-P1 : chicken /A 248 (Q90627) U15155 TRYlCHICK
rypsin 1: atlantic cod 3.4.21.4 241 (P16049) X76886 rypsin 1: human 3.4.21.4 247 47'UMAN

rypsin I, anionic: rat 3.4.21.4 246 (P00762) V01273 TRYIRAT
rypsin I: atlantic salmon 3.4.21.4 242 (P35031) X70075 iypsin: yellowfever mosquito 3.4.21.4 243 (P19799) X53458 rypsin 2: african malaria mosquito 3.4.21.4 277 (P35036) Z18890 trypsin, anionic: bovine 3.4.21.4 247 (Q29463) X54703 rypsin, anionic: dog 3.4.21.4 247 (P06872) M11589 ypsin I-P38 : chicken /A 248 (Q90628) U15156 TRY2CHICK.
trypsin II: human 3.4.21.4 247 (P07478) M27602 trypsin,I1, anionic: rat 3.4.21.4 246 (P00763) V01274 rypsin II: atlantic salmon 3.4.21.4 231 (P35032) X70073 rypsin: yellowfever mosquito N/A 244 (P70059) U72330 rypsin 3A1: yellowfever mosquito 3.4.21.4 254 (P29786) X64362 trypsin3: african malaria mosquito 3.4.21.4 275 (P35037) Z22930 trypsin II-P29 : chicken /A 248 (090629) U15157 rypsin III: human 3.4.21.4 247 (P15951) X15505 trypsin alpha-3: green-bottle fly, 3.4.21.4 165 (P35043) L15632 australian sheep blowfly TRY3LUCCU
trypsin III, cationic: rat 3.4.21.4 247 (P08426) M16624 trypsin. III: atlantic salmon 3.4.21.4 238 (1'35033) X70074 trypsin 4: african malaria mosquito 3.4.21.4 275 (P35038) Z2293D

trypsin 4A: human 3.4.21.4 304 (P35030) X72781 rypsin alpha-4: green-bottle fly, 3.4.21.4 (P35044) L15632 australian sheep blowfly 255 TRY4LUCCU
trypsin IV: rat 3.4.21.4 247 (P127$8) X15679 iypsin 5G1: yellowfever mosquito 3.4.21.4 238 (P29787) X64363 rypsin 5: african malaria mosquito 3.4.21.4 274 (P35039) Z22930 rypsin 6: african malaria mosquito 3.4.21.4 273 (P35040) Z22930 rypsin 7: african malaria mosquito 3.4.21.4 267 (P35041) Z22930 rypsin alpha:fruit fly 3.4.21.4 256 (P54624) U40653 TRYADROER
rypsin alpha: fruit fly 3.4.21.4 256 (P04814) X02989 TRYADROME
alpha-tryyptase: human 3.4.21.59 275 (P15157) M30038 TRYAHUMAN
rypsin, alkaline A: tobacco hawkmoth, N/A 256 (P35045) L16805 orwarm TRYAMANSE
rypsin V-A: rat 3.4.21.4 246 (P32821) X59012 TRYARAT
rypsin beta:fruit fly 3.4.21.4 253 (P54625) U40653 TRYBDROER

rypsin beta: fruit fly 3.4.21.4 253 (P35004) M96372 TRYBDROME
eta-tryptase: human 3.4.21.59 275 (P20231) M37488 TRYB HUMAN
trypsin, alkaline B: tobacco hawkmoth, tA 256 (P35046) L16806 orworm TRYBMANSE
rypsinV-B: rat 3.4.21.4 246 (P32822) X59013 TRYBRAT
trypsin, alkaline C : tobacco hawkmoth, (P35047) orworm rypsin delta/gamma: fruit fly 3.4.21.4 253 (P54626) U40653 TRYD DROER

trypsin delta: fruit fly 3.4.21.4 253 (P42276) U04853 TRYD DROME
rypsin epsilon: fruit fly 3.4.21.4 256 (P54627) U40653 TRYEDROER
trypsin epsilon: fruit fly 3.4.21.4 256 (P35005) M96372 TRYEDROME
trypsin gamma: fruit fly 3.4.21.4 253 (P42277) U04853 TRYGDROME
trypsin iota: fruit fly 3.4.21.4 252 (P52905) U41476 TRYIDROME
rypsin precursor: streptomyces griseus 3.4.21.- 259 (P00775) /A
TRYPSTRGR
ryptase-like protease: rat 3.4.21.59 23 (P27436) /A
TRYLRAT

astocytoma protease: dog 3.4.21.- 269 (P 19236) TRYMCANFA M24665 ast cell tryptase: rat 3.4.21.59 274 (P50343) D38455 TRYMRAT
rypsin I, : broad-fingered crayfish 3.4.21.4 237 (P00765) /A
TRYPASTFL
trypsin CFT-1: spruce budworm 3.4.21.4 256 (P35042) L04749 TRYPCHOFU
rypsin: cat 3.4.21.4 16 (P81071) jA
TRYPFELCA
rypsin: fusarium oxysporum 3.4.21.4 248 (P35049) S63827 TRYPFUS OX
rypsin: mouse 3.4.21.4 246 (P07146) X04574 TRYPMOUSE
rypsin: penoeid shrimp. 3.4.21.4 32 (P35050) fA
TRYPPENMO
rypsin: pig 3.4.21.4 231 (P00761) /A
TRYPPIG
trypsin: plaise 3.4.21.4 250 (P35034) X56744 TRYPPLEPL
trypsin: marbled lungfish 3.4.21.4 21 (P35051) /A
TRYPPROAT
rypsin: streptomyces erythraeus 3.4.21.4 227 (P24664) fA
TRYPSACER
rypsin: grey flesh fly 3.4.21.4 254 (P51588) X94691 TRYPSARBU
trypsin: black fly 3.4.21.4 247 (P35048) L08428 TRYPSIMVI
rypsin: spiny dogfish 3.4.21.4 229 (P00764) /A
TRYP_SQUAC
rypsin-like protease: streptomyces 3.4.21.- 268 (Q54179) U13770 glaucescens TRYP STRGA
, rypsin: streptomyces griseus 3.4.21.4 259 (P00775) M64471 TRYP STRGR

tryptase: dog 3.4.21.59 275 (P15944) M24664 TRYTCANFA
rypsin theta: fruit fly 3.4.21.4 262 (P54628) U40653 TRYTDROER
trypsin theta: fruit fly 3.4.21.4 262 (P42278) U04853 TRYTDROME

nast cell tryptase: mongolian jird 3.4.21.59 270 (P50342) D31789 TRYTMERUN
trypsineta: fruit fly 3.4.21.4 258 (P54629) U40653 TRYUDROER
trypsineta: fruit fly 3.4.21.4 262 (P42279) U04853 TRYUDROME

trYPsinX: atlantic cod 3.4.21.4 241 (Q91041) X76887 TRYXGADMO
rypsin zeta: fruit fly 3.4.21.4 281 (P54630) U40653 TRYZDROER
sin zeta: fruit fly 3.4.21.4 280 (P42280) U04853 TRYZDROME
rokinase-type plasmin activator: 3.4.21.73 433 (Q05589) L03546 ovine UROK BOVIN
rokinase-type plasmin activator: 3.4.21.73 434 (P15120) J05187 chicken UROKCHICK
rokinase-type plasmin activator: 3.4.21.73 431 (P00749) X02419 iuman UROKHUMAN
rokinase-type plasmin activator: 3.4.21.73 433 (P06869) X02389 ouse UROK MOUSE
rokinase-type plasmin activator: 3.4.21.73 433 (P16227) X51935 ellow baboon UROKPAPCY
rokinase-type plasmin activator: pig 3.4.21.73 442 (P04185) X01648 UROKPIG
rokinase-type plasmin activator: rat 3.4.21.73 432 (P29598) X63434 UROKRAT

issue plasmin activator: bovine 3.4.21.68 566 (Q28198) X85800 UROTBOVIN
tissue plasmin activator: human 3.4.21.68 562 (P00750) X07393 UROTHUMAN
issue plasmin activator: mouse 3.4.21.68 559 (P11214) J03520 UROT MOUSE

issue plasmin activator: rat 3.4.21.68 559 ( LJR
salivary plasmin activator alpha 1: 3.4.21.68 (P98119) M63987 ampire bat 477 URT1DESRO
salivary plasmin activator alpha 2: 3 (P15638) M63988 vampire bat .4.21.68 477 URT2DESRO
salivary plasmin activator beta: vampire 3.4.21.68 431 (P98121) M63989 bat URTB DESRO

saiivary piasmin aciivator gamma: 3.4.21.68 394 (P49150) M63990 ampire bat URTGDESRO
itellin-degradingprotease: silk moth 3.4.21.- 264 (Q07943) D16232 VDP BOMMO
5.2.2 Vectors Encoding a Protease [0083] Any suitable method known to one skilled in the art without limitation for expressing a heterologous gene in a cell can be used to express the heterologous protease or pro-protease. Typically, a recoinbinant nucleic acid construct comprising a nucleic acid encoding the protease or pro-protease is constructed using conventional molecular biology techniques, then the construct is introduced into a host cell line of interest. Cells comprising the desired construct are identified, then screened to identify cells that express the heterologous protein to the desired concentration. Methods for performing each of these operations are legion.

[0084] Numerous vectors suitable for this purpose are publicly available, including, but not limited to, plasmids, bacteriophage, viral vectors, retroviral vectors, artificial chromosomes and episomal vectors. Methods by which a construct can be selected and used such vectors are well known to those skilled in the art. Such vectors may be used for simple cloning and mutagenesis; however, gene expression vectors should be employed when introducing a nucleic acid encoding a protease or pro-protease into a suitable cell. The vector may be selected to accommodate a protease or pro-protease coding sequence of any desired size, typically from 0.25 kilobases (kb) to 40 kb or more in length.

[0085] Vectors typically contain various functional components, including a cloning (or "polylinker") site, an origin of replication and at least one selectable marker gene. Further, to express the protease or pro-protease, the vectors typically possess one or more of the following: enhancer element, promoter, transcription terinination and signal sequences, each positioned in the vicinity of the cloning site, such that they are operatively linked to the nucleic acid encoding a protease or pro-protease.

[0086] The expression of a protease or pro-protease protein may be controlled by any promoter or enhancer element known in the art. Suitable promoters which may be used include, but are not limited to, the SV40 early promoter region (Bemoist and Chambon, 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., 1980, Ce1122:787-797), the herpes thymidine kinase promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al., 1982, Nature 296:39-42), the tetracycline (Tet) promoter (Gossen et al., 1995, Proc. Nat. Acad. Sci. USA 89:5547-5551), or a CMV
promoter (such as the human immediate-early CMV promoter). Further, should temporal control of the expression of the protease or pro-protease be desirable, any inducible promoter known to one skilled in the art without limitation can be used according to the present invention. For example, the regulatory eleinent can be an interferon-responsive inducible proinoter or regulatory element. See, e.g., Levy et al., 1988, Gefzes & Devel.
2:383-393, Reich et al., 1987, P.N.A.S. USA 84:6394-6398, and Pellegrini et al., 1989, Mol Cell Biol.
9:4605-4612. Vectors may also be inducible because they contain horinone response elements, such as the glucocorticoid response element (GRE) and the estrogen response element (ERE), which can confer hormone inducibility where vectors are used for expression in cells having the respective hormone receptors. To reduce background levels of expression, elements responsive to ecdysone, an insect honnone, can be used instead, with coexpression of the ecdysone receptor.

[0087] Vectors generally contain nucleic acid sequences that enable the vector to replicate in one or more selected host cells. However, when the vector is intended to integrate into the host cell's genoine, the vector need not be able to autonoinously replicate, and, in fact, desirably does not do so. Typically, this sequence enables the vector to replicate independently of the host chromosomal DNA and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, viruses, and mammalian cells.

[0088] The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g. SV
40, adenovirus) are useful for cloning vectors in mammalian cells. Generally, the origin of replication is not needed for mainmalian expression vectors unless these are used in mammalian cells able to replicate high levels of DNA, such as COS cells.

[0089] Advantageously, the vector may contain a selection gene also referred to as selectable marker. This gene encodes a protein necessary for the survival or growth of transforined host cells grown in a selective culture mediuin. Host cells not transformed with the vector containing the selection gene will therefore not survive in the culture medium. Typical selection genes encode proteins that confer resistance to antibiotics and other toxins, e.g.
ampicillin, neomycin, methotrexate or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available in the growth media. Since the vectors encoding protease or pro-protease are most cominonly introduced into mammalian cells, a mammalian selectable marker, for example, a G418 resistance gene, can advantageously be einployed.
[0090] A number of selection systems may be used, including but not limited to the herpes simplex virus thyinidine kinase (Wigler et al., 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1962, Proc. Natl. Acad. Sci.
USA
48:2026), and adenine phosphoribosyltransferase (Lowy et al., 1980, Ce1122:817) genes can be employed in tk-, hgprt- or aprt-cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for dhfi, which confers resistance to methotrexate (Wigler et al., 1980, Natl. Acad. Sci. USA 77:3567; O'Hare et al., 1981, Proc. Natl. Acad.
Sci. USA
78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc.
Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., 1981, J. Mol. Biol. 150:1); and hygro, which confers resistance to hygromycin (Santerre et al., 1984, Gene 30:147) genes. Other antibiotic-resistance genes, such as those conferring resistance to ampicillin, Claforan, gentamycin, G41 8, hygromycin, rifampicin, kanamycin, neomycin, spectinomycin, or tetracycline, may also find use as selectable markers.

[0091] Expression vectors typically contain a promoter that is recognized by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive. The term "operably linked" refers to a juxtaposition wherein the components described are in a relationship pernzitting thein to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.

[0092] Construction of vectors encoding a protease or pro-protease employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and religated in the form desired to generate the required vector. If desired, analysis to confirin that the correct sequences are present in the constructed vector can be performed in a known fashion.
Suitable inethods for constructing expression vectors, preparing in vitro transcripts, introducing DNA into host cells, and performing analyses for assessing expression and function are known to those skilled in the art. The presence of a gene sequence in a sample is detected, or its ainplification and/or expression quantified by conventional methods, such as Southern or Northern analysis, Western blotting, dot blotting of DNA, RNA or protein, in situ hybridization, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.

[0093] In cases where an adenovirus is used as an expression vector, the protease or pro-protease coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chiineric gene may then be inserted in the adenovirus genoine by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the protease or pro-protease in infected hosts (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. Sci. USA 8 1:355-359).
Alternatively, a retroviral expression vector containing the Harvey murine sarcoma virus (Ha-MSV) long terminal repeats (LTRs) flanking the promoter and nucleic acid encoding the modified ABC transporter polypeptide may be used. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.

[0094] Artificial chromosomes may also be employed to deliver larger fragments of DNA
than can be contained in and expressed from a plasmid or retroviral vector.
Artificial chromosomes of about 6 kb to 10 Mb can be constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes. (See, e.g., Harrington, J. J. et al. (1997) Nat. Genet. 15:345-355.) [0095] Specific initiation signals may also be required for efficient translation of inserted protease or pro-protease coding sequences. These signals include the ATG
initiation codon and adjacent sequences. Furthermore, the initiation codon should be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be ei-i.ianced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc.
(see, e.g., Bittner et al., 1987, Methods in Enzymol. 153:516-544).

[0096] In certain embodiments, cells that stably express a protease or pro-protease are provided. To produce such cell lines, rather than using expression vectors that contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription tei-rninators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched medium, and then are switched to a selective medium. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasinid into their chromosomes and grow to fonn foci that in turn can be cloned and expanded into cell lines.
5.2.3 Methods for Making a Cell Expressing a Heterologous Protease [0097] The methods chosen for introducing the vector encoding a protease or pro-protease to a desired cell will typically depend on the vector and the cell.

[0098] Transfonnation and other methods of introducing nucleic acids into a host cell (e.g., conjugation, protoplast transfonnation or fusion, transfection, electroporation, liposome delivery, membrane fusion techniques, high velocity DNA-coated pellets, viral infection and protoplast fusion) can be accomplished by a variety of methods that are well known in the art (See, for instance, Ausubel, infra, and Sambrook et al., infra). Bacterial, yeast, plant or mainmal.ian cells can be transformed or transfected with an expression vector, such as a plasmid, a cosinid, or the like, wherein the expression vector coinprises the nucleic acid of interest, as described above. Alternatively, the cells may be infected by a viral expression vector comprising the nucleic acid of interest. Depending upon the host cell, vector, and method of transformation used, transient or stable expression of the polypeptide will be constitutive or inducible. One having ordinary skill in the art will be able to decide whether to express a polypeptide transiently or stably, and whether to express the protein constitutively or inducibly, as described above.

[0099] Mammalian cells can be directly infected by packaged viral vectors, or transfected by chemical or electrical means. For chemical transfection, DNA can be coprecipitated with CaPO4 or introduced using liposomal and nonliposomal lipid-based agents.
Corninercial kits are available for CaPO4 transfection (CalPhosTM Mammalian Transfection Kit, Clontech Laboratories, Palo Alto, Calif., USA), and lipid-mediated transfection can be practiced using commercial reagents, such as LIPOFECTAMIN RO 2000, LIPOFECTAMINETM Reagent, CELLFECTIN RO Reagent, LIPOFECTINO Reagent (Invitrogen, Carlsbad, Calif., USA), DOTAP Liposoinal Transfection Reagent, FuGENE 6, X-tremeGENE Q2, DOSPER, (Roche Molecular Biochemicals, Indianapolis, Ind. USA), EffecteneTM, PolyFectO, and Superfect (Qiagen, Inc., Valencia, Calif., USA). Protocols for electroporating mammalian cells can be found in, for example,; Norton et al. (eds.), Gene Transfer Methods:
Introducing DNA into Living Cells and Organisms, BioTechniques Books, Eaton Publishing Co. (2000).
Other transfection techniques include transfection by particle bombardment and microinj ection.

See, e.g., Cheng et al., Proc. Natl. Acad. Sci. USA 90(10): 4455-9 (1993);
Yang et al, Proc.
Natl. Acad. Sci. USA 87(24): 9568-72 (1990).

5.2.4 Culturing Cells Expressing a Heterologous Protease j01001 The cells expressing a heterologous protease can be cultured in any suitable culture medium known to one skilled in the art without limitation. Typically, cells are cultured in a standard cominercial culture medium, such as Dulbecco's modified Eagle's medium supplemented with sei-um (e.g., 10% fetal bovine serum), or in serum free medium, under controlled humidity and CO2 concentration suitable for maintaining neutral buffered pH (e.g., at pH between 7.0 and 7.2). Optionally, the medium contains antibiotics to prevent bacterial growth, e.g., penicillin, streptoznycin, etc., and/or additional nutrients, such as L-glutainine, sodiuin pyruvate, non-essential amino acids, additional supplements to promote favorable growth characteristics, e.g., trypsin, (3-mercaptoethanol, and the like.

[0101] Procedures for maintaining mammalian cells in culture have been extensively reported, and are known to those of skill in the art. General protocols are provided, e.g., in Freshney (1983) Culture of Animal Cells: Manual of Basic Technique, Alan R.
Liss, New York; Paul (1975) Cell and Tissue Culture, 5th ed., Livingston, Edinburgh;
Adams (1980) Laboratory Techniques in Biochemistry and Molecular Biology-Cell Culture for Biochemists, Work and Burdon (eds.) Elsevier, Amsterdam. Additional details regarding tissue culture procedures of particular interest in the production of influenza virus in vitro include, e.g., Merten et al. (1996) Production of influenza virus in cell cultures for vaccine preparation. In Cohen and Shafferman (eds) Novel Strategies in Design and Production of Vaccines, which is incorporated herein in its entirety. Additionally, variations in such procedures adapted to the present invention are readily determined through routine experimentation.

[01021 In one embodiment, the cells of the invention are cultivated as adherent cells on a surface to which they attach. Adherent surfaces on which tissue culture cells can be grown on are well known in the art. Adherent surfaces include but are not limited to, surface modified polystyrene plastics, protein coated surfaces (e.g., fibronectin and/or collagen coated glass/plastic) as well as a large variety of coznmercially available microcarriers (e.g., DEAE-Dextran microcarrier beads, such as Dormacell, Pfeifer & Langen;
Superbead, Flow Laboratories; styrene copolymer-tri-methylamine beads, such as Hillex, SoloHill, Ann Arbor).

[0103] In one embodiment, the cells of the invention are cultivated as suspension cells without a carrier. Methods for adapting cells to for growth in suspension without a carrier are known in the art (see, e.g., U.S. Patent Nos. 6,825036 and 6,455,298).
Alternatively, or optionally, the level of protease or pro-protease expressed by the cells of the invention may be regulated so as to result in the growth of the cells in suspension without any adaptation. In certain embodiments, the cells of the invention expression sufficient levels of protease or pro-protease to grow in suspension without adaptation. In other einbodiments, the cells of the invention grown as suspension cells without adaptation are used for the replication of viru.ses.
In a specific einbodiment, the cells of the invention grown as suspension cells without adaptation are used for the replication of influenza viruses.

[0104] In one embodiment, the cells of the invention grow as suspension cells without adaptation due to expression of a protease or pro-protease. In certain embodiments, the protease or pro-protease is a serine protease. In other embodiments, the protease or pro-protease is trypsin or trypsinogen. rn still other embodiments, the protease or pro-protease is a mammalian trypsin or trypsinogen. In yet other embodiments, the protease or pro-protease is a bacterial trypsin or tiypsinogen.

[0105] In some embodiments, the level of protease or pro-protease expressed by the cells of the invention growing in suspension is between about 0.1 ng and about 50 g per ml of cell culture. In other embodilnents, the level of protease or pro-protease expressed by the cells of the invention growing in suspension is at least 0.1 ng, or at least 0.5 ng, or at least 1.0 ng, or at least 5.0 ng, or at least 10 ng, or at least 20 ng, or at least 30 ng, or at least 40 ng, or at least 50 ng, or at least 60 ng, or at least 70 ng, or at least 80 ng, or at least 90 ng, or at least 1 g, or at least 2 g, or at least 5 g, or at least 10 g, or at least 20 g, or at least 30 g, or at least 40 g, or at least 50 g per ml of cell culture.

[0106] Cells for production of influenza virus can be cultured in serum-containing or seruin free medium. In some case, e.g., for the preparation of purified viruses, it is desirable to grow the host cells in serum free conditions. Appropriate serum fiee media are described in U.S. Provisional Application No. 60/638,166, filed Deceinber 23, 2004, U.S.
Provisional Application No. 60/641,139, filed January 5, 2005 and U.S. Patent Application No.
11/304,589, filed December 16, 2005, each of which is hereby incorporated by reference in its entirety.

[0107] It will be appreciated by one of skill in the art that the use of serum or animal extracts in tissue culture applications may have drawbacks (Lambert, K.J. et al., In:
Animal Cell Biotechnology, Vol 1, Spier, R.E. et al., Eds., Academic Pres New York, pp. 85-122 (1985)).
For example, the chemical composition of these supplements may vary between lots, even from a single manufacturer. In addition, suppleinents of animal or human origin may also be contaminated with adventitious agents (e.g., mycoplasma, viruses, and prions).
These agents can seriously undermine the health of the cultured cells when these contaminated supplements are used in cell culture media formulations. Fur-ther, these agents may pose a health risk when substances produced in cultures contaminated with adventitious agents are used in cell therapy and other clinical applications. A major fear is the presence of prions which cause spongiforin encephalopathies in animals and Creutzfeld-Jakob disease in humans. Thus, in certain embodiments, the culture media is completely serum free.
Advantageously, the culture medium can be completely free of animal products.
Accordingly, in certain embodiments, the culture media is animal protein free (APF). In certain embodiments, no exogenous animal-derived protease is added to the culture medium.
In, certain embodiments, no animal derived product is added to the culture medium. Specific media formulations are disclose in, for example, U.S. Patent Application No.
11/304,589, filed December 16, 2005.

[0108] Cells can be cultured in small scale, e.g., less than 25 ml medium, culture tubes or flasks or in large flasks with agitation, in rotator bottles, or on microcarrier beads (e.g., DEAE-Dextran microcarrier beads, such as Donnacell, Pfeifer & Langen;
Superbead, Flow Laboratories; styrene copolymer-tri-methylamine beads, such as Hillex, SoloHill, Ann Arbor) in flasks, bottles or reactor cultures. Microcarrier beads are small spheres (in the range of 100-200 microns in diameter) that provide a large surface area for adherent cell growth per voluine of cell culture. For example a single liter of medium can include more than 20 million microcarrier beads providing greater than 8000 square centiineters of growth surface.
For commercial production of viruses, e.g., for vaccine production, it is often desirable to culture the cells in a bioreactor or fermenter. Bioreactors are available in volumes froin t under 1 liter to in excess of 1001iters, e.g., Cyto3 Bioreactor (Osmonics, Minnetonka, MN);
NBS bioreactors (New Brunswick Scientific, Edison, N.J.); laboratory and commercial scale bioreactors from B. Braun Biotech International (B. Braun Biotech, Melsungen, Germany).
[0109] Regardless of the culture volume, in certain embodiments, it is important that the cultures be maintained at a temperature less than or equal to 35 C, to insure efficient recovery of recombinant and/or reassortant influenza virus that are to some extent cold-adapted. For example, in certain embodiments, the cells are cultured at a temperature between about 32 C and 35 C, typically at a temperature between about 32 C
and about 34 C, usually at about 33 C.

[0110] Typically, a regulator, e.g., a thermostat, or other device for sensing and maintaining the temperature of the cell culture systein can be employed to insure that the temperature does not exceed 35 C during the period of virus replication.

5.3 SPRT Protease from StNeptorrzyces griseus [0111] In addition to the proteases described above, the present invention provides a novel bacterial protease, termed SPRT. The gene encoding this protease was cloned out of Streptomyces griseus as described in the examples below. The SPRT protease is suitable for expressing in cells to be used for culturing viruses, as described above.

[0112] The nucleotide sequence of the sprT gene is presented as Figure 9 (SEQ
ID NO:1), while the amino acid sequence of the SPRT protease is presented as Figure 10 (SEQ ID
NO:2). In addition to the native nucleic acid encoding the sprT gene, nucleic acids encoding sequences homologous to the nucleic acid sequence of the sprT gene are also conteinplated according to the present invention. Thus, in certain embodiinents, the invention provides a nucleic acid encoding a nucleotide sequence that is about 99%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, or about 50%
identical to the nucleic acid sequence of Figure 9. In another aspect, the invention provides, a nucleic acid that encodes a polypeptide that has a sequence that is about 99%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, or about 50% identical to the sequence of the polypeptide encoded by the nucleic acid sequence of Figure 9.

[0113] In another embodiment, the invention provides a nucleic acid that hybridizes to a nucleic acid comprising the nucleotide sequence presented as Figure 9 (or encoding a polypeptide of Table 1). In certain embodiments, the nucleic acid hybridizes to the nucleic acid comprising the nucleotide sequence presented as Figure 9 under defined hybridization conditions. In certain embodiments, the hybridization conditions are stringent hybridization conditions. In certain embodiments, the hybridization conditions are highly stringent hybridization conditions.

[0114] Furthermore, the nucleic acids of the invention also encompass derivative versions of nucleic acids encoding an SPRT protease. Such derivatives can be made by any method known by one of skill in the art without limitation. For example, derivatives can be made by site-specific mutagenesis, including substitution, insertion, or deletion of one, two, three, five, ten or more nucleotides, of the nucleic acids. Alternatively, derivatives can be made by random mutagenesis. One method for randomly mutagenizing a nucleic acid coinprises amplifying the nucleic acid in a PCR reaction in the presence of 0.1 mM MnC12 and unbalanced nucleotide concentrations. These conditions increase the inisincorporation rate of the polymerase used in the PCR reaction and result in random mutagenesis of the amplified nucleic acid.

[0115] In certain embodiments, the derivative nucleic acids encoding derivatives of the SPRT
protease have improved properties relative to the wild-type enzyme described herein. For example, in some embodiments, the derivative nucleic acids encode a derivative SPRT
protease that has a greater activity, e.g., greater specific activity than the wild-type enzyme.
In some embodiments, the derivative SPRT protease can have a derivative secretion signal that increases secretion of the SPRT protease relative to the wild-type protease. In soine embodiments, the derivative SPRT protease can have a derivative prepropeptide sequence that increases cleavage of the prepropeptide from the SPRT protease relative to the wild-type protease. In some embodiments, the derivative SPRT protease exhibits an activity maximum at a different pH from that of the wild-type protease. In certain embodiments, the pH is the pH preferred for culturing viruses, e.g., influenza viruses.

[0116] In other aspects, the invention provides a SPRT polypeptide. In certain embodiments, the amino acid sequence of the SPRT polypeptide is at least about 99%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, or about 50% identical to the ainino acid sequence of Figure 10. In other aspects, the invention provides a protein having an ainino acid sequence of the polypeptide that is at least about 99%, about 95%, about 90%, about 85%, about 80%, about 75%, about 70%, about 65%, about 60%, about 55%, or about 50% identical to the ainino acid sequence of Table 1.
Further, the invention provides active fragments of a SPRT polypeptide or protease of the invention. In certain embodiments, the active fragments comprise at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, or 320 contiguous amino acids selected from the amino acid sequence of Figure 10 (or Table 1) while retaining protease activity. The skilled artisan can routinely identify such active fragments by, for example, expressing the fraginent and testing for protease activity using conventional techniques.

[0117] Accordingly, in another aspect, the present invention relates to artificial variants coinprising a conservative substitution, deletion, and/or insertion of one or more amino acids of the mature polypeptide of SEQ ID NO:2 (or Table 1); or a homologous sequence thereof.
In a specific embodiment, the amino acid changes are of a minor nature, that is conservative amino acid substitutions or insertions that do not significantly affect the folding and/or activity of the protein; small deletions, typically of one to about 30 amino acids; small amino-or carboxyl-terminal extensions, such as an ainino-tenninal methionine residue; a small linker peptide of up to about 20-25 residues; or a small extension that facilitates purification by changing net charge or another function, such as a poly-histidine tract, an antigenic epitope or a binding domain.

[0118] Examples of conservative substitutions are within the group of basic amino acids (arginine, lysine and histidine), acidic amino acids (glutamic acid and aspartic acid), polar amino acids (glutamine and asparagine), hydrophobic amino acids (leucine, isoleucine and valine), aromatic amino acids (phenylalanine, tryptophan and tyrosine), and small amino acids (glycine, alanine, serine, threonine and methionine). Amino acid substitutions which do not generally alter specific activity are known in the art and are described, for example, by H.
Neurath and R. L. Hill, 1979, In, The Proteins, Academic Press, New York. The most commonly occurring exchanges are Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Tyr/Phe, Ala/Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu, and Asp/Gly.

[0119] In addition to the 20 standard amino acids, non-standard amino acids (such as 4-hydroxyproline, 6-M-methyl lysine, 2-aminoisobutyric acid, isovaline, and alpha-methyl serine) may be substituted for amino acid residues of a wild-type polypeptide.
A limited number of non-conseivative amino acids, amino acids that are not encoded by the genetic code, and unnatural amino acids may be substituted for amino acid residues.
"Unnatural amino acids" have been inodified after protein synthesis, and/or have a chemical structure in their side chain(s) different from that of the standard amino acids. Unnatural amino acids can be chemically synthesized, and are commercially available, and include pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, and 3,3-dimethylproline.

[0120] Alternatively, the ainino acid changes are of such a nature that the physico-cheinical properties of the polypeptides are altered. For example, amino acid changes inay improve the thermal stability of the polypeptide, alter the substrate specificity, change the pH optimum, and the like.

[0121] Essential amino acids in the parent polypeptide can be identified according to procedures known in the art, such as site-directed inutagenesis or alanine-scanning mutagenesis (Cunningham and Wells, 1989, Science 244: 1081-1085). In the latter technique, single alanine mutations are introduced at every residue in the molecule, and the resultant mutant molecules are tested for biological activity (i.e., lipase activity) to identify amino acid residues that are critical to the activity of the molecule. See also, Hilton et al., 1996, J. Biol.
Chem. 271: 4699-4708. The active site of the enzyme or other biological interaction can also be determined by physical analysis of structure, as determined by such techniques as nuclear magnetic resonance, crystallography, electron diffraction, or photoaffinity labeling, in conjunction with mutation of putative contact site amino acids. See, for example, de Vos et al., 1992, Science 255: 306-312; Smith et al., 1992, J. Mol. Biol. 224: 899-904; Wlodaver et al., 1992, FEBS Lett. 309: 59-64. The identities of essential amino acids can also be inferred from analysis of identities with polypeptides which are related to a polypeptide according to the invention.

[0122] Single or multiple amino acid substitutions can be made and tested using known methods of inutagenesis, recombination, and/or shuffling, followed by a relevant screening procedure, such as those disclosed by Reidhaar-Olson and Sauer, 1988, Science 241: 53-57;
Bowie and Sauer, 1989, Proc. Natl. Acad. Sci. USA 86: 2152-2156; WO 95/17413;
or WO
95/22625. Other methods that can be used include error-prone PCR, phage display (e.g., Lowman et al., 1991, Biochem. 30: 10832-10837; U.S. Pat. No. 5,223,409; WO
92/06204), and region-directed inutagenesis (Derbyshire et al., 1986, Gene 46: 145; Ner et al., 1988, DNA 7: 127).

[0123] Mutagenesis/shuffling methods can be combined with high-throughput, automated screening methods to detect activity of cloned, inutagenized polypeptides expressed by host cells (Ness et al., 1999, Nature Biotechnology 17: 893-896). Mutagenized DNA
molecules that encode active polypeptides can be recovered from the host cells and rapidly sequenced using standard methods in the art. These inethods allow the rapid determination of the iinportance of individual amino acid residues in a polypeptide of interest, and can be applied to polypeptides of unknown structure.

5.4 Expression Vectors [0124] In still another aspect, the invention provides expression vectors for expressing the SPRT protease or another protease of the invention (See, e.g., Table 1).
Generally, expression vectors are recombinant polynucleotide molecules comprising expression control sequences operatively linked to a nucleotide sequence encoding a polypeptide. Expression vectors can readily be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, selectable markers, etc. to result in stable transcription and translation of mRNA. Techniques for construction of expression vectors and expression of genes in cells coinprising the expression vectors are well known in the art.
See, e.g., Sambrook et al., 2001, Molecular Cloning -- A Labot=atozry Manual, 3d edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, and Ausubel et al., eds., Current Edition, Curt-ent Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.

[0125] Useful promoters for use in expression vectors include, but are not limited to, a metallothionein promoter, a constitutive adenovirus major late promoter, a dexamethasone-inducible MMTV promoter, a SV40 promoter, a MRP pol III promoter, a constitutive MPSV
promoter, a tetracycline-inducible CMV promoter (such as the human immediate-early CMV
promoter), a constitutive CMV promoter, and an interferon-responsive promoter.

[0126] The expression vectors should contain expression and replication signals compatible with the cell in which the SPRT protease is expressed. Suitable expression vectors include, but are not limited to, viral vectors such as retroviruses, adenoviruses and adenoassociated viruses, plasmid vectors, cosmids, and the like. Viral and plasmid vectors are preferred for transfecting the expression vectors into mammalian cells. For example, the expression vector pcDNAl (Invitrogen, San Diego, CA), in which the expression control sequence coinprises the CMV promoter, provides good rates of transfection and expression into such cells.
Further examples of expression vectors that can be used are provided in the Examples, below.
[0127] The expression vectors can be introduced into a cell by any method known to one of skill in the art without liinitation. Such methods include, but are not limited to, e.g., direct uptake of the molecule by a cell from solution; facilitated uptake through lipofection using, e.g., liposomes or immunoliposomes; particle-mediated transfection; etc. See, e.g., U.S.
Patent No. 5,272,065; Goeddel et al., eds, 1990, Methods in Enzynaology, vol.
185, Academic Press, Inc., CA; Krieger, 1990, Gene Transfer and Expressiofz -- A Laboratofy Manual, Stockton Press, NY; Sambrook et al., 1989, Molecular Cloning -- A Laboratofy Manual, Cold Spring Harbor Laboratory, NY; and Ausubel et al., eds., Current Edition, Curt=ent Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.
[0128] The expression vectors can also contain a purification moiety that simplifies isolation of the delivery construct. For exainple, a polyhistidine moiety of, e.g., six histidine residues, can be incorporated at the amino terminal end of the protein. The polyhistidine moiety allows convenient isolation of the protein in a single step by nickel-chelate chromatography. In certain embodiments, the purification moiety can be cleaved from the remainder of the delivery construct following purification. In other einbodiments, the moiety does not interfere witli the function of the functional domains of the delivery construct and thus need not be cleaved.

5.5 Additional Methods for Manipulation of Nucleic Acids and Proteins [0129] In the context of the invention, nucleic acids, including viral nucleic acids, nucleic acids encoding a protease or pro-protease, and the like, can be manipulated according to well known molecular biology techniques. Detailed protocols for numerous such procedures, including amplification, cloning, mutagenesis, transformation, and the like, are described in, e.g., in Ausubel et al. Current Protocols in Molecular Biology (supplemented through 2006) John Wiley & Sons, New York ("Ausubel"); Sainbrook et al. Molecular Cloning -A
Laboratory Manual (3rd Ed.), Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 2001 ("Sambrook"), and Berger and K.immel Guide to Molecular Cloning Techniques, Methods in Enzymolo~y volume 152, Academic Press, Inc., San Diego, CA
("Berger").

[0130] In addition to the above references, protocols for in vitro ainplification techniques, such as the polymerase chain reaction (PCR), the ligase chain reaction (LCR), Q(3-replicase amplification, and other RNA polymerase mediated techniques (e.g., NASBA), useful e.g., for amplifying cDNA probes of the invention, are found in Mullis et al. (1987) U.S. Patent No. 4,683,202; PCR Protocols A Guide to Methods and Applications (Innis et al.
eds) Academic Press Inc. San Diego, CA (1990) ("Innis"); Arnheim and Levinson (1990) C&EN
36; The Journal Of NIH Research (1991) 3:81; Is'-woh et al. (1989) Proc Natt Acad Sci USA
86, 1173; Guatelli et al. (1990) Proc Natl Acad Sci USA 87:1874; Lomell et al.
(1989) J Clin Chein 35:1826; Laildegren et al. (1988) Science 241:1077; Van Brunt (1990) Biotechnology 8:291; Wu and Wallace (1989) Gene 4: 560; Barringer et al. (1990) Gene 89:117, and Sooknanan and Malek (1995) Biotechnology 13:563. Additional methods, useful for cloning nucleic acids in the context of the present invention, include Wallace et al.
U.S. Pat. No.

5,426,039. hnproved methods of amplifying large nucleic acids by PCR are suinmarized in Cheng et al. (1994) Nature 369:684 and the references therein.

[0131] Certain polynucleotides of the invention, e.g., oligonucleotides can be synthesized utilizing various solid-phase strategies including mononucleotide- and/or trinucleotide-based phosphoramidite coupling chemistry. For example, nucleic acid sequences can be synthesized by the sequential addition of activated monomers and/or trimers to an elongating polynucleotide chain. See e.g., Caruthers, M.H. et al. (1992) Meth Enzpnol 211:3.

[0132] In lieu of synthesizing the desired sequences, essentially any nucleic acid can be custom ordered from any of a variety of coinmercial sources, such as The Midland Certified Reagent Company, The Great American Gene Company, ExpressGen, Inc., Operon Technologies, Inc., and many others.

[0133] In addition, substitutions of selected amino acid residues in viral polypeptides, proteases, or pro-proteases, can be accomplished by, e.g., site directed mutagenesis. For example, viral polypeptides with amino acid substitutions functionally correlated with desirable phenotypic characteristic, e.g., an attenuated phenotype, cold adaptation, temperature sensitivity, cleavage by a particular protease, etc. can be produced by introducing specific mutations into a viral nucleic acid segment encoding the polypeptide.
Methods for site directed mutagenesis are well known in the art, and described, e.g., in Ausubel, Sambrook, and Berger, supra. Numerous kits for performing site directed mutagenesis are commercially available, e.g., the Chameleon Site Directed Mutagenesis Kit (Stratagene, La Jolla), and can be used according to the manufacturers instructions to introduce, e.g., one or more amino acid substitutions into a genome segment encoding a influenza A or B
polypeptide, respectively, or into a nucleic acid encoding a protease or pro-protease.

5.6 Use of Cells Expressing a Heterologous Protease to Culture Influenza [0134] Methods for replicating influenza viruses in cell culture are known to one of skill in the art (See, section entitled "Culturing Cells Expressing a Heterologous Protease," supra).
Typically, these methods involve the infection of suitable host cells with a selected strain of virus. Alternatively, recombinant methods are used to introduce the viral genome into the host (e.g., plasmid rescue detailed in section entitled "Influenza Genoinic Vectors in Host Cells Expressing a Protease or Pro-Protease," itafra). As discussed extensively above, exogenous proteases have typically been added to culture medium to efficiently produce influenza viruses in cell culture for cells that do not express a protease that efficiently cleaves the HAO protein (see, e.g., Appleyard, et al., 1974, J.Gen Virol. 25:351-357;
U.S. Patents 5,824,536; 4,500,513; and Patent Publication WO 96/15232). One of the objects of the present invention is to provide a cell that expresses a heterologous protease to replicate influenza, and in a specific embodiment, improve the efficiency of influenza infection to increase viral titers in cell culture.

[0135] Accordingly, in certain einbodiments, exogenous protease, e.g., porcine trypsin, is not added to the cell culture medium in which viruses, e.g., influenza viruses, are to be replicated.
In certain embodiments, the titer of the influenza virus yielded from a cell culture expressing a heterologous protease without exogenous protease is equal or substantially equal to the titer that would be achieved from a cell culture not expressing a heterologous protease with the addition of exogenous protease. In other embodiments, the titer of the influenza virus yielded from a cell culture expressing a heterologous protease without exogenous protease is greater than the titer that would be achieved from a cell culture not expressing a heterologous protease with the addition of exogenous protease. In still otller embodiments, a cell culture expressing a heterologous protease is capable of propagating influenza virus to a commercially reasonable titer (>107 Log TCID50/mL).

[0136] In yet other embodiments, exogenous protease, e.g., porcine trypsin or SPRT
protease, can be added to a cell culture comprising cells of the invention in which viruses, e.g., influenza viruses, are to be replicated. Such embodiments are useful, for example, to culture viruses in cells of the invention that express a protease at low levels, e.g., levels insufficient to propagate the virus to commercially reasonable titer. Such embodiments are also useful in methods where the cells of the invention expresses a pro-protease that is activated by proteolytic cleavage.

[0137] In one embodiment, methods of generating infectious viral particles of a negative-strand RNA virus in cultured cells are provided, wherein no exogenous protease, e.g., trypsin, is added to the cell culture medium. In a specific embodiment, methods of replicating negative-strand RNA viruses to a titer (logio TCID50/mL) of at least about 6.0, or at least 6.2, or at least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8 in a population of cells is provided, wherein no exogenous protease is added to the cell culture medium.

[0138] In certain embodiments, the titer (logio TCID50/mL) of influenza virus yielded from a cell culture of cells expressing a heterologous protease without exogenous protease is at least 6.0, or at least 6.2, or at least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8.

[0139] In a specific embodiment, the titer of the influenza virus yielded from a cell culture of cells expressing a heterologous protease without exogenous protease has a logio TCID50/mL
that is at least about 0.1, or at least about 0.2, or at least about 0.3, or at least about 0.4, or at least about 0.5, or at least about 0.6, or at least about 0.7, or at least about 0.8, or at least about 0.9, or at least about 1.0, or at least about 1.2, or at least about 1.4, or at least about 1.6, or at least about 1.8, or at least about 2.0, or at least about 2.2, or at least about 2.4, or at least about 2.6, or at least about 2.6, or at least about 2.8, or at least about 3.0, or at least about 3.2, or at least about 3.4, or at least about 3.6, or at least about 3.8, or at least about 4.0, or at least about 4.2, or at least about 4.4, or at least about 4.6, or at least about 4.8, or at least about 5.0, greater then the titer of influenza virus produced in a culture of corresponding cells that do not express a heterologous protease and to which no exogenous protease, e.g., trypsin has been added.

[0140] In other einbodiments, less protease than would otherwise be added to the cell culture medium can be added to the cell culture medium. Accordingly, in certain embodiments, methods of generating infectious viral particles of a negative-strand RNA
virus in cultured cells are provided, wherein a minimal ainount of exogenous protease, e.g., trypsin, is added to the cell culture medium are provided. In a specific embodiment, methods of replicating negative-strand RNA viruses to a titer (loglo TCID50/mL) of at least about 6.0, or at least 6.2, or at least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8 in a population of cells are provided, wherein about 0.1 ng/nll to about 100 g/ml exogenous protease is added to the cell culture medium.

[0141] In another a specific embodiment, methods of replicating negative-strand RNA
viruses to a titer (loglo TCID50/mL) of at least about 6.0, or at least 6.2, or a:t least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8 in a population of cells are provided, wherein about 1 mU/ml to about 5000 mU/ml exogenous protease is added to the cell culture medium.

[0142] In still another embodiment, the titer (logio TCID50/mL)of influenza virus yielded from a cell culture of cells expressing a heterologous protease, to which about 0.1 ngfml to about 100 ttg/ml of exogenous protease has been added, is at least 6.0, or at least 6.2, or at least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8. In a specific embodiment, the titer (logio TCID50/mL)of influenza virus yielded from a cell culture of cells expressing a heterologous protease, to which about 0.1 ng/ml to about 10 g/ml of exogenous protease has been added, is at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8. In another specific embodiment, the titer (loglo TCID50/mL)of influenza virus yielded from a cell culture of cells expressing a heterologous protease, to which about 0.1 ng/ml to about 1.0 g/ml of exogenous protease has been added, is at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8.

[0143] In certain embodiments, the titer (logio TCID50/mL)of influenza virus yielded from a cell culture of cells expressing a heterologous protease, to which about 1 mU/ml to about 5000 mU/ml of exogenous protease has been added, is at least 6.0, or at least 6.2, or at least 6.4, or at least 6.6, or at least 6.8, or at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8. In a specific einbodiment, the titer (loglo TCID50/mL)of influenza virus yielded from a cell culture of cells expressing a heterologous protease, to which about 1 mU/ml to about 1000 mU/ml of exogenous protease has been added, is at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8. In another specific embodilnent, the titer (logio TCID50/mL)of influenza virus yielded from a cell culture of cells expressing a heterologous protease, to which about 1 mU/ml to about 500 mU/ml of exogenous protease has been added, is at least 7.0, or at least 7.2, or at least 7.4, or at least 7.6, or at least 7.8, or at least 8.0, or at least 8.2, or at least 8.4, or at least 8.6, or at least 8.8, or at least 9.0 , or at least 9.2, or at least 9.4, or at least 9.6, or at least 9.8.

[0144] In certain embodiments, the titer of virus is obtained after incubation of the cells for about about 2 days to about 10 days, or optionally about 3 days to about 7 days. In a specific embodiment, the titer of virus is obtained after incubation of the cells for 2 days, or 3 days, or 4 days, or 5 days, or after 6 days, or 7 days, or 8 days, or 9 days, or 10 days, or 12 days, or 14 days.

[0145] In certain einbodiments, exogenous protease is added to the cell culture medium to a final concentration of less than about 0.1 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 0.1 ng/ml. In certain einbodiments, exogenous protease is added to the cell culture medium to a final concentration of about 0.5 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 1 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 5 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 10 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 50 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 100 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 250 ng/ml. In certain embodiments, exogenous protease is added to the cell culture mediuin to a final concentration of about 500 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 750 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 1 g/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 5 g/m1. In certain embodiments, exogenous protease is added to the cell culture inedium to a final concentration of about 10 g/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration of about 25 g/ml. In certain einbodiments, exogenous protease is added to the cell culture medium to a final concentration of about 50 gg/ml. In certain embodiments, exogenous protease is added to the cell culture mediuin to a final concentration of about 100 g/ml.

[0146] In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 0.01 ng/ml and about 100 g/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 1 ng/ml and about 100 g/ml. In certain embodiments, exogenous protease is added to the cell culture mediuin to a final concentration between about 10 ng/ml and about 100 gg/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 100 ng/inl and about 100 gg/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 1 g/ml and about 100 g/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 10 gg/ml and about 100 g/ml.

[0147] In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 0.01 ng/ml and about 10 g/mi. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 0.01 ng/ml and about 1 g/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a fmal concentration between about 0.01 ng/ml and about 100 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 0.01 ng/ml and about 10 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 0.01 ng/ml and about 1 ng/ml. In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 0.01 ng/ml and about 0.1 ng/ml.

[0148] In certain embodiments, exogenous protease is added to the cell culture medium to a final concentration between about 1 to 5000 mU/ml, or 5 to 1000 inU/ml, or 100 to 500 mU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a final concentration of less than about 1 mU/ml. In one einbodiinent, exogenous protease is added to the cell culture medium to a final concentration of less than about 5 inU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a final concentration of less than about 10 mU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a:final concentration of less than about 25 mU/ml. In one einbodiinent, exogenous protease is added to the cell culture inedium to a final concentration of less than abou't 50 mU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a final concentration of less than about 100 inU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a final concentration of less than about 250 mU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a final concentration of less than about 500 mU/ml. In one embodiment, exogenous protease is added to the cell culture medium to a final concentration of less than about 1000 mU/ml.

[0149] These embodiments where less protease than would typically be required for efficient production of influenza virus is added to the culture mediuin are particularly useful when the cell cultured expresses a pro-protease. Addition of the exogenous protease to the culture medium can "prime" the pro-protease by cleaving off the protective pro-peptide, converting the pro-protease into its active form. Thereafter, the activated protease can cleave pro-proteases subsequently expressed by the cell, thereby converting these newly produced pro-proteases to active proteases. Such embodiments thus. allow greatly reduced usage of exogenously added proteases, such as porcine trypsin.

[0150] Accordingly, in certain embodiments, exogenous protease is added to the cell culture in a single application. In certain einbodiments, the exogenous protease is added at or near the same time that the virus is used to infect the cells in the cell culture.
In certain embodiments, the exogenous protease is added on the same day that the virus is used to infect the cells in the cell culture. In other embodiments, the exogenous protease is added prior to the addition of the virus used to infect the cells in the cell culture. In certain embodiments, the exogenous protease is added at or near the same time that vectors are introduced into the cells in the cell culture for production of the virus. In certain embodiments, the exogenous protease is added on the same day that vectors are introduced into the cells in the cell culture for production of the virus. In other einbodiments, the exogenous protease is added prior to the addition of the vectors that are introduced into the cells in the cell culture for production of the virus.

[0151] Alternatively, in certain embodiments, the cell can express both a protease and a pro-protease. In certain of such embodiments, expression of the protease can be under the control of an inducible promoter to permit the transient expression of the protease.
Such einbodiments allow the protease to be temporarily expressed, thereby activating the pro-protease. Thereafter, the activated pro-protease can continue to activate subsequently expressed pro-protease. Alternately, in certain embodiinents, the cell can express two or more proteases or pro-proteases. In certain embodimetns, the two or lnore pro-proteases can be expressed under the saine or different regulatory systems, e.g., consitutive or inducible expression. For inducible expression embodiments, the inducible systemselected can be the same or different for each inducibly expressable protease or pro-protease.

[0152] In certain einbodiments, it is useful to temporally control the expression of the heterologous protease in the cell culture. For example, it may prove useful to induce expression of the protease at a defined time, e.g., about 1, 2, 3, 4, or 5 days following infection of the cell culture with an influenza virus. In other embodiments, the expression of the protease is induced prior to infection of the cell culture with an influenza virus. In still other embodiments, the protease is induced at the same time as infection of the cell culture with an influenza virus In yet other einbodiinents, the protease is induced prior to, at the same time or after the addition of the vectors that are introduced into the cells in the cell culture for production of the virus. Thus, in certain embodiments, the heterologous protease can be under the control of an inducible promoter or otherwise under the control of a genetic regulatory element, as described above.

5.7 Influenza Genomic Vectors in Host Cells Expressing a Protease or Pro-Protease [0153] In addition to cell culture-based methods that rely on infecting the cell culture with live virus, fully infectious influenza viruses can be produced in cell culture using recombinant DNA technology, e.g. plasmid rescue. See, e.g., Neumann et al. (1999) Genes ation of influenza A virus entirely f om cloned cDNAs. Proc Natl Acad Sci USA 96:9345-9350; Fodor et al. (1999) Rescue of influenza A viy-us ft=om recombinant DNA. J. Virol 73:9679-9682;
Hoffinann et al. (2000) A DNA transfection system fot generation of influenza A virus from eight plasmids Proc Natl Acad Sci USA 97:6108-6113; WO 01/83794; Hoffinann and Webster (2000), Unidirectional RNA polymef ase I-polymerase II transcyiption svstemfoY the generation of influenza A virus f om eight plasmids, 81:2843-2847; Hoffinann et al. (2002), Rescue of influenza B virfuses frotn 8 plastnids, 99(17): 11411-11416; U.S.
patent nos.
6,649,372 and 6,951,754; U.S. publication nos. 20050003349 and 20050037487, which are incorporatated by reference herein.

[0154] In certain embodiments, the methods of the invention comprise introducing a plurality of vectors, each of which encodes a portion of an influenza virus genome into cells expressing a heterologous protease or pro-protease to obtain influenza viruses. The cells expressing a heterologous protease or pro-protease are then cultured under conditions permissive for viral growth, and influenza viruses are recovered. In sozne embodiments, the influenza viruses are attenuated viruses, cold adapted viruses and/or temperature sensitive viruses. For example, in certain embodiments, the vector-derived recoinbinant influenza viruses can be attenuated, cold adapted, teinperature sensitive viruses, such as are suitable for adininistration as a live attenuated vaccine, e.g., in a intranasal vaccine fonnulation. In an exemplary embodiment, the viruses are produced by introducing a plurality of vectors incorporating all or part of an influenza B/Ann Arbor/1/66 virus genome, e.g., a ca B/Ann Arbor/1/66 virus genome.

[0155] In some embodiments, a plurality of vectors coinprising cDNA encoding at least the 6 internal genome segments of one influenza strain and eDNA encoding one or more genome segments (e.g., HA and NA vRNA segments) of a different influenza strain can be introduced into cells expressing a heterologous protease or pro-protease to obtain influenza viruses. For example, at least the 6 internal genome segments ("the backbone") of an attenuated, cold adapted and/or temperature sensitive influenza A or B strain, e.g., a ca, att, ts strain of B/Ann Arbor/1/66, can be introduced into cells expressing a heterologous protease or pro-protease along with one or more segments encoding iminunogenic antigens derived from another virus strain. Typically the immunogenic surface antigens include either or both of the hemagglutinin (HA) and/or neuraminidase (NA) antigens. In embodiments where a single segment encoding an immunogenic surface antigen is introduced, the 7 complementary segments of the selected virus are also introduced into the host cells.

[0156] In some embodiments, a plurality of vectors comprising cDNA encoding 1-7 internal genome seginents of one influenza strain and cDNA encoding 1-7 genome segments (e.g., HA and NA vRNA segments) of a different influenza strain can be introduced into cells expressing a heterologous protease or pro-protease to obtain influenza viruses.

[0157] In certain embodiments, expression vectors encoding influenza vRNA are cotransfected into the cells expressing a heterologous protease or pro-protease by electroporation. In certain embodiments, the expression vectors are introduced into cells expressing a heterologous protease or pro-protease by transfection into cells in the presence of a liposoinal transfection reagent or by means of calcium phosphate precipitation. In certain einbodiments, the expression vectors are plasmids. In certain embodiments, the expression vectors comprise a separate expression vector for expression of each genomic RNA segment of said virus or the corresponding coding RNAs.

[0158] In certain embodiments, a plurality of plasmid vectors encoding each influenza virus vRNA are introduced into a population of host cells. For example, in certain embodiment, 8 plasmids, each of which encodes a different vRNA segment can be utilized to introduce a complete influenza genome into the host cells. In one einbodiment, plasmid vectors also encode mRNA of at least one influenza polypeptide. Alternatively, a greater number of plasmids, incorporating smaller genomic subsequences can be einployed.

[0159] In accordance with the present invention, viral genomic RNA
corresponding to each of the eight genomic segments of influenza can be inserted into a recoinbinant expression vector for manipulation and production of influenza viruses. A variety of vectors, including viral vectors, plasmids, cosmids, phage, and artificial chromosomes, can be employed in the context of the invention. Typically, for ease of manipulation, the viral genomic segments are inserted into a plasmid vector, providing one or more origins of replication functional in bacterial and eukaryotic cells, and, optionally, a marker convenient for screening or selecting cells incorporating the plasmid sequence. These vectors can then be introduced into cells expressing a heterologous protease or pro-protease to obtain influenza viruses.

[0160] In another embodiment, the invention provides a method for producing a recombinant influenza virus, comprising introducing into cells of the invention a plurality of expression vectors comprising an RNA pol I promoter operably linked to one or more cI?NAs encoding each influenza genomic RNA and one or more expression vectors that express viral mRNA
that encodes one or more influenza polypeptides: PB2, PB1, PA, HA, NP, NA, M1, M2, and NS2; and isolating, i.e., harvesting said recombinant influenza virus from the cells.

[0161] In one einbodiment, the present invention provides for methods of generating infectious recombinant influenza virus in host cells of the invention using expression vectors to express the vRNA seginents or corresponding cRNAs and influenza virus proteins, in particular PB1, PB2, PA and NA. In accordance with this embodiment, helper virus may or may not be included to generate the infectious recombinant influenza viruses.

[0162] The present invention provides a method for generating in cultured cells of the invention infectious viral particles of a negative-strand RNA virus, said method comprising:
(a) introducing into a population of said cells a set of expression vectors capable of both expressing in said cells genoinic vRNA to provide the coinplete genomic vRNA
of said virus and capable of expressing mRNA encoding one or more polypeptides of said virus; (b) culturing said cells whereby said viral particles are produced. In certain embodiments, the cells are canine cells. In certain embodiments, the cells are MDCK cells. In certain einbodiments, the virus is influenza B virus. In certain einbodiinents, the set of expression vectors is contained in 1-17 plasmids. In certain embodiments, the set of expression vectors is contained in 1-8 plasmid. In certain embodiments, the set of expression vectors is contained in 1-3 plasmids. In certain embodiments, the sets of expression vectors are introduced by electroporation. In certain embodiments, the set of expression vectors encode each vRNA segment of an influenza virus. In certain embodiments, the set of expression vectors encode the mRNA of one or more influenza polypeptide. In certain einbodiiuents, the set of expression vectors encode each vRNA seginent of an influenza virus and the mRNA of one or more influenza polypeptide. In certain einbodiments, the set of expression vectors encode a vRNA or mRNA of a second virus. For instance, the set of vectors comprises one or more vectors encoding the HA and/or NA mRNA and/or vRNA of a second influenza virus. In one embodiment, helper virus is used in the method. In one embodiment, the cultured cells used in the method are canine cells.

[0163] The present invention further provides a method for generating in cultured cells of the invention infectious recombinant viral particles of a negative-strand RNA
virus, said method comprising: (a) introducing into a population of said cells a first set of expression vectors capable of expressing in said cells genomic vRNA to provide the complete genomic vRNA of said virus; (b) introducing into said cells a second set of expression vectors capable of expressing mRNA encoding one or more polypeptides of said virus; and (c) culturing said cells whereby said viral particles are produced. In certain embodiments, the cells are canine cells. In certain embodiments, the cells are MDCK cells. In certain embodiments, the virus is influenza B virus. In certain einbodiments, the first set of expression vectors is contained in 1-8 plasmids. In certain embodiinents, the first set of expression vectors is contained in one plasmid. In certain embodiments, the second set of expression vectors is contained in 1-8 plasmids. In certain embodiments, the second set of expression vectors is contained in one plasmid. In certain embodiments, the first, second, or both sets of expression vectors are introduced by electroporation. In certain einbodiments, the first set of expression vectors encode each vRNA segment of an influenza virus. In certain embodiments, the second set of expression vectors encode the mRNA of one or more influenza polypeptide. In certain embodiments, the first set or second set of expression vectors (or both sets) coinprise a nucleic acid of the invention. In one embodiment, helper virus is used in the method.

[0164] The present invention also provides a method for generating in cultured cells of the invention infectious recoinbinant viral particles of a seginented negative-strand RNA virus having greater than 3 genomic vRNA segments, for example an influenza virus such as an influenza A virus, said method comprising: (a) introducing into a population of said cells a first set of expression vectors capable of expressing in said cells genomic vRNA segments to provide the complete genomic vRNA seginents of said virus; (b) introducing into said cells a second set of expression vectors capable of expressing mRNA encoding one or more polypeptides of said virus; and (c) culturing said cells whereby said viral particles are produced. In certain embodiments, the cells are canine cells. In certain einbodiments, the cells are MDCK cells. In certain embodiments, the recombinant virus is influenza A or B
virus. hi certain embodiments, the first set of expression vectors is contained in 1-8 plasinids.
In certain einbodiments, the first set of expression vectors is contained in one plasinid. In certain embodiments, the second set of expression vectors is contained in 1-8 plasmids. In certain embodiments, the second set of expression vectors is contained in one plasmid. In certain embodiments, the first, second, or both sets of expression vectors are introduced by electroporation. In certain einbodiments, the first set of expression vectors encode each vRNA segment of an influenza virus. In certain embodiments, the second set of expression vectors encode the mRNA of one or more or all influenza polypeptides. In certain embodiments, the first set or second set of expression vectors (or both sets) encode a vRNA
or mRNA of a second virus. For instance, a set of vectors comprises one or more vectors encoding the HA and/or NA mRNA and/or vRNA of a second influenza virus. In one embodiment, helper virus is used in the method.

[0165) The present invention further provides a method for generating in cultured cells of the invention infectious recombinant viral particles of a segxnented negative-strand RNA virus having greater than 3 genomic vRNA seginents, for exainple an influenza virus such as an influenza A virus, said method comprising: (a) introducing into a population of said cells a set of expression vectors capable of both expressing in said cells genomic vRNA segments to provide the complete genomic vRNA segments of said virus and capable of expressing mRNA encoding one or more polypeptides of said virus; (b) culturing said cells whereby said viral particles are produced. In certain embodiments, the cells are canine cells. In certain embodiments, the cells are MDCK cells. In certain embodiments, the virus is influenza A or B virus. In certain embodiments, the set of expression vectors is contained in 1-17 plasmids.
In certain embodiments, the set of expression vectors is contained in 1-8 plasmid. In certain embodiments, the set of expression vectors is contained in 1-3 plasmids. In certain embodiments, the set of expression vectors is contained in one plasmid. In certain embodiments, the sets of expression vectors are introduced by electroporation.
In certain embodiments, the set of expression vectors encode each vRNA segment of an influenza virus.
In certain einbodiments, the set of expression vectors encode the mRNA of one or more influenza polypeptide. In certain einbodiments, the set of expression vectors encode each vRNA segment of an influenza virus and the mRNA of one or more influenza polypeptide.
In certain embodiments, the set of expression vectors comprise a nucleic acid of the invention. In certain embodiments, the set of expression vectors encode a vRNA
or inRNA of a second virus. For instance, the set of vectors coinprises one or more vectors encoding the HA and/or NA mRNA and/or vRNA of a second influenza virus. In certain einbodiments, the first set or second set of expression vectors (or both sets) encode a vRNA
or mRNA of a second virus. For instance, a set of vectors comprises one or more vectors encoding the HA
and/or NA mRNA and/or vRNA of a second influenza virus. In one embodiment, helper virus is used in the method.

[0166] The plasmid expression vectors may be bi-directional expression vectors capable of initiating transcription of the inserted viral genomic segment in either direction, that is, giving rise to both (+) strand and (~ ) strand viral RNA molecules. To effect bi-directional transcription, each of the viral genomic seginents is inserted into a vector having at least two independent promoters, such that copies of viral genomic RNA are transcribed by a first RNA polymerase promoter (e.g., a RNA pol I promoter), from one strand, and viral mRNAs are synthesized from a second RNA polymerase promoter (e.g., a RNA Pol II
promoter or other promoter that can initiate transcription by RNA pol II in cells).
Accordingly, the two proinoters can be arranged in opposite orientations flanking at least one cloning site (i.e., a restriction enzyme recognition sequence) preferably a unique cloning site, suitable for insertion of viral genomic RNA segments. Alternatively, an "ambisense" vector can be employed in which the (+) strand inRNA and the (-) strand viral RNA (as a cRNA) are transcribed from the same strand of the vector.

[0167] To ensure the correct 3' end of each expressed vRNA or cRNA, each vRNA
or cRNA
expression vector can incorporate a ribozyme sequence or appropriate terminator sequence downstream of the RNA coding sequence. This may be, for example, the hepatitis delta virus genomic ribozyme sequence or a functional derivative thereof, or the murine rDNA
tenninator sequence (Genbank Accession Number M12074). Alternatively, for example, a Pol I terminator may be employed (Neumann et al., 1994, Virology 202:477-479).
The RNA
expression vectors may be constructed in the saine manner as the vRNA
expression vectors described in Pleschka et al., 1996, J. Virol. 70:4188-4192; Hoffinann and Webster, 2000, J.
Gen Virol. 81:2843-2847; Hoffinann et al., 2002, Vaccine 20:3165-3170; Fodor et al., 1999, J. Virol. 73:9679-9682; Neumann et al., 1999, P.N.A.S.USA 96:9345-9350; and Hoffinann et al., 2000, Virology 267:310-317; U.S. patent nos. 6,649,372 and 6,951,754;
U.S. publication nos. 20050003349 and 20050037487, each of which is hereby incorporated by reference in its entirety.

[0168] In other systeins, the viral sequences transcribed by the pol I and pol II promoters can be transcribed from different vectors. In these embodiments, vectors encoding each of the viral genomic segments under the control of a pol I promoter and vectors encoding at least PA, PB 1, PB2, and NP under the control of a pol II promoter can be used.

[0169] In either case, with regard to the pol II promoter, the influenza virus genome segment to be expressed can be operably linked to an appropriate transcription control sequence (promoter) to direct mRNA synthesis. A variety of promoters are suitable for use in expression vectors for regulating transcription of influenza virus genome seginents. In certain embodiments, the cytomegalovirus (CMV) DNA dependent RNA Polymerase II
(Pol II) promoter is utilized. If desired, e.g., for regulating conditional expression, other promoters can be substituted which induce RNA transcription under the specified conditions, or in the specified tissues or cells. Numerous viral and mammalian, e.g., huinan promoters are available, or can be isolated according to the specific application contemplated. For example, alternative promoters obtained from the genomes of animal and huinan viruses include such promoters as the adenovirus (such as Adenovirus 2), papilloma virus, hepatitis-B virus, and polyoma virus, and various retroviral promoters. Mammalian promoters include, among many others, the actin promoter, immunoglobulin promoters, heat-shock,promoters, and the like. In a specific embodiment, the regulatory sequence comprises the adenovirus 2 major late promoter linked to the spliced tripartite leader sequence of human adenovirus 2, as described by Berg et al., Bio Techniques 14:972-978. In addition, bacteriophage promoters can be employed in conjunction with the cognate RNA polyinerase, e.g., the T7 promoter.

[0170] Expression vectors used to express viral proteins, in particular viral proteins for RNP
complex formation, will preferably express viral proteins hoinologous to the desired virus.
The expression of viral proteins by these expression vectors inay be regulated by any regulatory sequence known to those of skill in the art. The regulatory sequence may be a constitutive promoter, an inducible promoter or a tissue-specific promoter.

[0171] Further examples of promoters which may be used to control the expression of viral proteins in protein expression vectors include, but are not limited to, the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., 1980, Cell 22:787-797), the herpes thyinidine kinase promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci.
USA 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al., 1982, Nature 296:39-42); prokaryotic expression vectors such as the (3-lactamase promoter (Villa-Kainaroff et al., 1978, Proc. Natl. Acad. Sci. USA 75:3727-3731), or the tac promoter (DeBoer et al., 1983, Proc. Natl. Acad. Sci. USA 80:21-25); see also "Useful proteins from recombinant bacteria" in Scientific American, 1980, 242:74-94; plant expression vectors comprising the nopaline synthetase promoter region (Herrera-Estrella et al., Nature 303:209-213) or the cauliflower mosaic virus 35S RNA promoter (Gardner et al., 1981, Nucl. Acids Res. 9:2871), and the promoter of the photosynthetic enzyme ribulose biphosphate carboxylase (Herrera-Estrella et al., 1984, Nature 310:115-120); promoter elements from yeast or other fungi such as the Gal 4 promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, alkaline phosphatase promoter, and the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells (Swift et al., 1984, Cel138:639-646; Omitz et al., 1986, Cold Spring Harbor Symp. Quant.
Biol. 50:399-409; MacDonald, 1987, Hepatology 7:425-515); insulin gene control region which is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115-122), iinmunoglobulin gene control region which is active in lymphoid cells (Grosschedl et al., 1984, Cell 38:647-658; Adames et al., 1985, Nature 318:533-538; Alexander et al., 1987, Mol. Cell. Biol. 7:1436-1444), mouse maminary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., 1986, Ce1145:485-495), albumin gene control region which is active in liver (Pinkert et al., 1987, Genes and Devel. 1:268-276), alpha-fetoprotein gene control region which is active in liver (Kruinlauf et al., 1985, Mol. Cell. Biol. 5:1639-1648; Hammer et al., 1987, Science 235:53-58; alpha 1-antitrypsin gene control region which is active in the liver (Kelsey et al., 1987, Genes and Devel. 1:161-171), beta-globin gene control region which is active in myeloid cells (Mogram et al., 1985, Nature 315:338-340; Kollias et al., 1986, Ce1146:89-94; myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-712), myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-286), and gonadotropic releasing hormone gene control region which is active in the hypothalainus (Mason et al., 1986, Science 234:1372-1378).

[0172] In a specific embodiinent, the protein expression vector comprises a promoter operably linked to a nucleic acid sequence, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene). In another embodiment, a protein expression vector that is capable of producing bicistronic mRNA may be produced by inserting bicistronic mRNA sequence. Certain internal ribosome entry site (IRES) sequences may be utilized. Preferred IRES elements include, but are not liinited to the mammalian BiP IRES and the hepatitis C virus IRES.

[0173] Expression vectors containing gene inserts can be identified, e.g., by three general approaches: (a) nucleic acid hybridization; (b) presence or absence of "marker" gene functions; and (c) expression of inserted sequences. In the first approach, the presence of the viral gene inserted in an expression vector(s) can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to the inserted gene(s).
In the second approach, the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., resistance to antibiotics or transformation phenotype) caused by the insertion of the gene(s) in the vector(s). In the third approach, expression vectors can be identified by assaying the gene product expressed. Such assays can be based, for example, on the physical or functional properties of the viral protein in in vits'o assay systeins, e.g., binding of viral proteins to antibodies.

[0174] In a specific embodiment, one or more protein expression vectors encode and express the viral proteins necessary for the fonnation of RNP coinplexes. In another embodiment, one or more protein expression vectors encode and express the viral proteins necessary to form viral particles. In yet another embodiment, one or more protein expression vectors encode and express the all of the viral proteins of a particular negative-strand RNA
virus.

[0175] Transcription can optionally be increased by including an enhancer sequence.
Enhancers are typically short, e.g., 10-500 bp, cis-acting DNA elements that act in concert with a promoter to increase transcription. Many enhancer sequences have been isolated from mammalian genes (hemoglobin, elastase, albuinin, alpha.-fetoprotein, and insulin), and eukaryotic cell viruses. The enhancer can be spliced into the vector at a position 5' or 3' to the heterologous coding sequence, but is typically inserted at a site 5' to the promoter.
Typically, the promoter, and if desired, additional transcription enhancing sequences are chosen to optimize expression in the host cell type into which the heterologous DNA is to be introduced (Scharf et al. (1994) Heat stress promoters and transcription factors Results Probl Cell Differ 20:125-62; Kriegler et al. (1990) Assenably of enhancef s, promoters, and splice signals to control expression of transfef-red genes Methods in Enzymol 185:
512-27).
Optionally, the ainplicon can also contain a ribosome binding site or an internal ribosoine entry site (IRES) for translation initiation.

[0176] The vectors can also include sequences for the terinination of transcription and for stabilizing the mRNA, such as a polyadenylation site or a terminator sequence.
Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. In some embodiments, the SV40 polyadenylation sequences provide a polyadenylation signal.

[0177] In addition, as described above, the expression vectors optionally include one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells, in addition to genes previously listed, markers such as dihydrofolate reductase or neomycin resistance are suitable for selection in eukaryotic cell culture.

[0178] The vector containing the appropriate DNA sequence as described above, as well as an appropriate proinoter or control sequence, can be employed to transform a host cell permitting expression of the protein. While the vectors can be replicated in bacterial cells, most frequently it will be desirable to introduce them into mammalian cells, e.g., Vero cells, BHK cells, MDCK cell, 293 cells, COS cells, for the purpose of expression. In a specific embodiment, MDCK cells are used for the purpose of expression.

[0179] The expression vectors can be used to directing the expressing of genomic vRNA(s) or corresponding cRNA(s) which have one or more inutations. These mutations may result in the attenuation of the virus. For example, the vRNA segments may be the vRNA
segments of an influenza A virus having an attenuated base pair substitution in a pan-handle duplex promoter region, in particular, for example, the known attenuating base pair substitution of A
for C and U for G at position 11-12' in the duplex region of the NA-specific vRNA (Fodor et al., 1998, J. Virol. 6923-6290). By using the methods to produce recoinbinant negative-strand RNA viras, new attenuating mutations may be identified.

[01801 Further, any of the expression vectors described in U.S. Patent Nos.
6,951,754, 6,887,699, 6,649,372, 6,544,785, 6,001,634, 5,854,037, 5,824,536, 5,840,520, 5,820,871, 5,786,199, and 5,166,057 and U.S. Patent Application Publication Nos.
20060019350, 20050158342,20050037487,20050266026,20050186563,20050221489,20050032043, 20040142003, 20030035814, and 20020164770 can be used in accordance with the present invention.

[0181] Still further, the expression vectors can also be used to make chimeric viruses that express sequences heterologous to a viral genome. Expression vectors directing the expression of vRNA(s) or corresponding cRNA(s) and introduced into host cells along with expression vectors direct the expression of viral proteins to generate novel infectious recoinbinant negative-strand RNA viruses or chimeric viruses. Heterologous sequences which may be engineered into these viruses include antisense nucleic acids and nucleic acid such as a ribozyme. Alternatively, heterologous sequences which express a peptide or polypeptide may be engineered into these viruses. Heterologous sequences encoding the following peptides or polypeptides may be engineered into these viruses include: 1) antigens that are characteristic of a pathogen; 2) antigens that are characteristic of autoiminune disease; 3) antigens that are characteristic of an allergen; and 4) antigens that are characteristic of a tumor. For example, heterologous gene sequences that can be engineered into the chimeric viruses of the invention include, but are not limited to, epitopes of human immunodeficiency virus (HIV) such as gp160; hepatitis B virus surface antigen (HBsAg); the glycoproteins of herpes virus (e.g., gD, gE); VP 1 of poliovirus; and antigenic deterniinants of nonviral pathogens such as bacteria and parasites to name but a few.

[0182] Yet further, the methods of the present invention may be modified to incorporate aspects of methods known to those skilled in the art, in order to improve efficiency of rescue of infectious viral particles. For example, the reverse genetics technique involves the preparation of synthetic recoinbinant viral RNAs that contain the non-coding regions of the negative strand virus RNA which are essential for the recognition by viral polymerases and for packaging signals necessary to generate a mature virion. The recombinant RNAs are synthesized from a recombinant DNA template and reconstituted in vitro with purified viral polymerase complex to form recombinant ribonucleoprotein (RNPs) which can be used to transect cells. A more efficient transfection is achieved if the viral polymerase proteins are present during transcription of the synthetic RNAs either in vitro or in vivo.
The synthetic recombinant RNPs can be rescued into infectious virus particles. The foregoing techniques are described in U.S. Pat. No. 5,166,057 issued Nov. 24, 1992; in U.S. Pat.
No. 5,854,037 issued Dec. 29, 1998; in U.S. Pat. No. 5,789,229 issued Aug. 4, 1998; in European Patent Publication EP 0702085A1, published Feb. 20, 1996; in U.S. Pat. application Ser. No.
09/152,845; in International Patent Publications PCR W097/12032 published Apr.
3, 1997;
W096/34625 published Nov. 7, 1996; in European Patent Publication EP-A780475;
W099/02657 published Jan. 21, 1999; W098/53078 published Nov. 26, 1998;

published Jan. 22, 1998; W099/15672 published Apr. 1, 1999; W098/13501 published Apr.
2, 1998; W097/06720 published Feb. 20, 1997; and EPO 780 47SA1 published Jun.
25, 1997, each of which is incorporated by reference herein in its entirety.

[0183] Vectors comprising influenza genome segments can be introduced (e.g., transfected) into host cells of the invention according to methods well known in the art for introducing heterologous nucleic acids into eukaryotic cells, including, e.g., calcium phosphate co-precipitation, electroporation, microinjection, lipofection, and transfection employing polyaiuine transfection reagents. For example, vectors, e.g., plasmids, can be transfected into host cells, such as COS cells, 293T cells, MDCK cells, or combinations of COS
or 293T cells and MDCK cells, using the polyamine transfection reagent TransIT-LT1 (Mirus) according to the manufacturer's instructions. Approximately 1 g of each vector to be introduced into the population of host cells with approximately 2 l of TransIT-LT1 diluted in 160 gl medium, (e.g., serum-free medium), in a total vol. of 200 l. The DNA:transfection reagent mixtures are incubated at room teinperature for 45 min followed by addition of 800 gl of medium. The transfection mixture is added to the host cells, and the cells are cultured as described above.
Accordingly, for the production of recoiubinant or reassortant viruses in cell culture, vectors incorporating each of the 8 genoiue segments, (PB2, PB 1, PA, NP, M, NS, HA
and NA) are mixed with approximately 20 l TranslT-LT1 and transfected into host cells.
Optionally, sei-u.m-containing medium is replaced prior to transfection with serum-free medium, e.g., Opti-MEM I, and incubated for 4-6 hours.

[0184] Alternatively, electroporation can be employed to introduce vectors incorporating influenza genome seginents into host cells of the invention. For exainple, plasmid vectors incorporating an influenza A or influenza B virus are favorably introduced into Vero cells using electroporation accordingto the following procedure. In brief, 5 x 106 Vero cells, e.g., grown in Modified Eagle's Mediuin (MEM) supplemented with 10% Fetal Bovine Serum (FBS) are resuspended in 0.4 ml OptiMEM and placed in an electroporation cuvette. Twenty micrograms of DNA in a volume of up to 25 l is added to the cells in the cuvette, which is then mixed gently by tapping. Electroporation is performed according to the manufacturer's instructions (e.g., BioRad Gene Pulser II with Capacitance Extender Plus connected) at 300 volts, 950 microFarads with a time constant of between 28-33 msec. The cells are remixed by gently tapping and approximately 1-2 minutes following electroporation 0.7 ml MEM
with 10% FBS is added directly to the cuvette. The cells are then transferred to two wells of a standard 6 well tissue culture dish containing 2 ml MEM, 10% FBS or OPTI-MEM
without seruin. The cuvette is washed to recover any remaining cells and the wash suspension is divided between the two wells. Final volume is approximately 3.5 mls. The cells are then incubated under conditions pennissive for viral growth, e.g., at approxiinately 33 C for cold adapted strains.

5.8 Recovery of Viruses from Cell Culture [0185] Viruses can be typically recovered from the culture mediuin, in which infected (transfected) cells have been grown. Typically crude medium is clarified prior to concentration of influenza viruses. Common metllods include filtration, ultrafiltration, adsorption on barium sulfate and elution, and centrifugation. For example, crude mediuin from infected cultures can first be clarified by centrifugation at, e.g., 1000-2000 x g for a time sufficient to remove cell debris and other large particulate matter, e.g., between 10 and 30 minutes. Alternatively, the mediuin is filtered through a 0.8 m cellulose acetate filter to remove intact cells and other large particulate matter. Optionally, the clarified medium supernatant is then centrifuged to pellet the influenza viruses, e.g., at 15,000 x g, for approximately 3-5 hours. Following resuspension of the virus pellet in an appropriate buffer, such as STE (0.01 M Tris-HCI; 0.15 M NaCI; 0.0001 M EDTA) or phosphate buffered saline (PBS) at pH 7.4, the virus is concentrated by density gradient centrifugation on sucrose (60%-12%) or potassiuin tartrate (50%-10%). Either continuous or step gradients, e.g., a sucrose gradient between 12% and 60% in four 12% steps, are suitable. The gradients are centrifuged at a speed, and for a time, sufficient for the viruses to concentrate into a visible band for recovery. Alternatively, and for most large scale commercial applications, virus is elutriated from density gradients using a zonal-centrifuge rotor operating in continuous mode.
Additional details sufficient to guide one of skill through the preparation of influenza viruses from tissue culture are provided, e.g., in Furminger. Vaccine Production, in Nicholson et al.
(eds) Textbook of Influenza pp. 324-332; Merten et al. (1996) Production of influenza virus in cell cultures for vacciiae preparation, in Cohen & Shafferman (eds) Novel Strategies in Design and Production of Vaccines pp. 141-151, and United States patents no.
5,690,937. If desired, the recovered viruses can be stored at -80 C in the presence of sucrose-phosphate-glutainate (SPG) as a stabilizer.

[0186] In one embodiment, the invention provides coinpositions comprising viruses (or portions thereof) replicated in the cells of the invention that have been treated with an agent such as benzonase; to eliminate potential oncogenes. Accordingly, an oncogene-free vaccine coinposition is specifically included within the embodiinents of the invention.
5.9 Other Viruses [0187] In addition to the influenza viruses described above, the cells and methods of the invention can also be used to culture other viruses. In certain einbodiments, one or more polypeptides of the virus undergo proteolytic cleavage by a host cell protease at some point during the life-cycle of the virus. In specific embodiments, use of a cell or method of the invention will increase the yield of the virus, the infectivity of the virus, or improve another desirable biological property of the virus known to one skilled in the art.

[0188] Accordingly, in cei-tain embodiments, the cultured virus is a DNA
virus. In certain embodiments, the virus is an RNA virus. In certain embodiments, the virus is a single-stranded DNA virus. In certain embodiments, the virus is a double-stranded DNA
virus. In certain embodiments, the virus is a positive-sense single-stranded RNA virus.
In certain einbodiments, the virus is a negative-sense single-stranded RNA virus. In certain embodiments, the virus is a double-stranded RNA virus. In certain embodiments, the virus is a reverse-transcribing virus.

[0189] In certain einbodiments, the virus is a,member of an order that is selected from the group consisting of Caudovirales, Mononegavirales, and Nidovirales.

[0190] In certain embodiments, the virus is a member of a family or subfainily that is selected from the group consisting of Myoviridae, Siphoviridae, Podoviridae, Rudiviridae, Tectiviridae, Corticoviridae, Lipothrixviridae, Plasmaviridae, Fuselloviridae, Phycodnaviridae, Guttaviridae, Poxviridae, Chordopoxvirinae, Entomopoxvirinae, Iridoviridae, Polydnaviridae, Herpesviridae, Alphaherpesvirinae, Betaherpesvirinae, Gammaherpesvirinae, Polyomaviridae, Papillomaviridae, Adenoviridae, Ascoviridae, Baculoviridae, Nimaviridae, and Asfarviridae.

[0191] In certain einbodiments, the virus is a member of a genus that is selected from the group consisting of T4-like viruses, P 1-like viruses, P2-like viruses, Mu-like viruses, SPO1-like viruses, (DH-like viruses, ?,-like viruses, T1-like viruses, T5-like viruses, c2-like viruses, L5-like viruses, WM1-like viruses, T7-like viruses, cp29-like viruses, P22-like viruses, Rudivirus, Tectivirus, Corticovirus, Alphalipothrixvirus, Betalipotluixvirus, Gainmalipothrixvirus, Plasmavirus, Fusellovirus, Chlorovirus, Prasinovirus, Prymnesiovirus, Phaeovirus, Raphidovirus, Coccolithovirus, Guttavirus, Orthopoxvirus, Parapoxvirus, Avipoxvirus, Capripoxvirus, Leporipoxvirus, Suipoxvirus, Molluscipoxvirus, Yatapoxvirus, Alphaentoinopoxvirus, Betaentomopoxvirus, Gammaentomopoxvirus, Iridovirus, Chloriridovirus, Ranavirus, Lyinphocystivirus, Ichnovirus, Bracovirus, Ictalurivirus, Simplexvirus, Varicellovirus, Mardivirus, Iltovirus, Cytomegalovirus, Muromegalovirus, Roseolovirus, Lymphocryptovirus, Rhadinovirus, Polyomavirus, Papillomavirus, Mastadenovirus, Aviadenovirus, Atadenovirus, Siadenovirus, Ascovirus, Mimivirus, Nucleopolyhedrovirus, Granulovirus, Whispovirus, Asfivirus, and Rhizidiovirus.
[0192] In certain embodiments, the virus is selected from the group consisting of Enterobacteria phage T4, Enterobacteria phage P 1, Enterobacteria phage P2, Enterobacteria phage Mu, Bacillus phage SP01, Halobacterium virus (DH, Enterobacteria phage k, Enterobacteria phage T1, Enterobacteria phage T5, Lactococcus phage c2, Mycobacterium phage L5, Methanobacterium VM1, Enterobacteria phage T7, Bacillus phage cp29, Enterobacteria phage P22, Sulfolobus virus SIRV1, Enterobacteria phage PRD1, Alteromonas phage PM2, Thermoproteus virus 1, Sulfolobus islandicus filamentous virus, Acidianus filainentous virus 1, Acholeplasma phage L2, Sulfolobus virus SSV1, Paramecium bursaria Chlorella virus 1, Micromonas pusilla vii-us SP'l, Chrysochroinulina brevifilum virus PW1, Ectocarpus siliculosis virus 1, Heterosigma akashiwo virus 01, Einiliania huxleyi virus 86, Sulfolobus neozealandicus droplet-shaped virus, Vaccinia virus, Orf virus, Fowlpox virus, Sheeppox virus, Myxoma virus, Swinepox virus, Molluscum contagiosum virus, Yaba monkey tuinor virus, Melolontha melolontha entoinopoxvirus, Amsacta inoorei entomopoxvirus, Chironomus luridus entomopoxvirus, Invertebrate iridescent virus 6, Invertebrate iridescent virus 3, Frog virus 3, Lymphocystis disease virus 1, Campoletis sonorensis ichnovirus, Cotesia melanoscela bracovirus, Ictalurid herpesvirus 1, Human herpesvirus 1, Huinan herpesvirus 3, Gallid herpesvirus 2, Gallid herpesvirus 1, Human herpesvirus 5, Murid herpesvirusl, Human herpesvirus 6, Human herpesvirus 4, Saimiriine herpesvirus 2, Simian virus 40, Cottontail rabbit papillomavirus, Human adenovirus C, Fowl adenovirus A, Ovine adenovirus D, Turkey adenovirus B, Spodoptera frugiperda ascovirus, Acanthamoeba polyphaga mimivirus, Autographa califomica nucleopolyhedrovirus, Cydia pomonella granulovirus, White spot syndrome virus 1, African swine fever virus, and Rhizidiomyces virus.

[0193] In certain embodiments, the virus is a member of a family or subfamily that is selected from the group consisting of Inoviridae, Microviridae, Geininiviridae, Circoviridae, Nanoviridae, Parvoviridae, Parvovirinae, and Densovirinae.

[0194] In certain embodiments, the virus is a member of a genus that is selected from the group consisting of Inovirus, Plectrovirus, Microvirus, Spiromicrovirus, Bdellomicrovirus, Chlamydiamicrovirus, Mastrevirus, Curtovirus, Begomovirus, Circovirus, Gyrovirus, Nanovirus, Babuvirus, Parvovirus, Erythrovirus, Dependovirus, Densovirus, Iteravirus, and Brevidensovirus.

[0195] In certain embodiments, the virus is selected from the group consisting of Enterobacteria phage M13, Acholeplasma phage MV-L51, Enterobacteria phage cpX174, Spiroplasma phage 4, Bdellovibrio phage MACI, Chlamydia phage 1, Maize streak virus, Beet curly top virus, Bean golden mosaic viius - Puerto Rico, Porcine circovirus, Chicken anaemia virus, Subterranean clover stunt virus, Babana bunchy top virus, Minute virus of mice, B 19 virus, Adeno-associated virus 2, Junonia coenia densovirus, Bombyx mori densovirus, and Aedes aegypti densovirus.

[0196] In certain embodiments, the virus is a member of a family or subfamily that is selected from the group consisting of Hepadnaviridae and Caulimoviridae.

[0197] In certain embodiments, the virus is a member of a genus that is selected from the group consisting of Orthohepadnavirus, Avihepadnavirus, Badnavirus, Caulimovirus, Tungrovirus, Soymovirus, Cavemovirus, and Petuvirus.

[0198] In certain embodiments, the virus is selected from the group consisting of Hepatitis B
virus, Duck hepatitis B virus, Commelina yellow mottle virus, Cauliflower mosaic virus, Rice tungro bacilliform virus, Soybean chlorotic mottle virus, Cassava vein mosaic virus, and Petunia vein clearing virus.

[0199] In certain embodiments, the virus is a member of a fainily or subfamily that is selected from the group consisting of Pseudoviridae, Metaviridae, and Retroviridae.
[0200] In certain embodiments, the virus is a member of a genus that is selected from the group consisting of Pseudovirus, Hemivirus, Metavirus, Errantivirus, Alpharetrovirus, Betaretrovirus, Gammaretrovirus, Deltaretrovirus, Epsilonretrovirus, Lentivirus, and Spumavirus.

[0201] In certain embodiments, the virus is selected from the group consisting of Saccharomyces cerevisiae Tyl virus, Drosophila melanogaster copia virus, Saccharomyces cerevisiae Ty3 virus, Drosophila melanogaster gypsy virus, Avian leucosis virus, Mouse mammary tumour virus, Murine leukeamia virus, Bovine leukaemia virus, Walleye dermal sarcoma virus, Human immunodeficiency virus 1, and Chimpanzee foamy virus.

[0202] In certain embodiments, the virus is a member of a fainily or subfamily that is selected from the group consisting of Cystoviridae, Reoviridae, Bimaviridae, Totiviridae, Chrysoviridae, Partitiviridae, and Hypoviridae.

[0203] In certain embodiments, the virus is a meinber of a genus that is selected from the group consisting of Cystovirus, Orthoreovirus, Orbivirus, Rotavirus, Coltivirus, Aquareovirus, Cypovirus, Fijivirus, Phytoreovirus, Oryzavirus, Aquabirnavirus, Avibimavirus, Entomobimavirus, Totivirus, Giardiavirus, Leishmaniavirus, Chrysovirus, Partitivirus, Alphacryptovirus, Betacryptovirus, Hypovirus, and Varicosavirus.
[0204] In certain embodiments, the virus is selected from the group consisting of Pseudomonas phage W6, Mammalian orthoreovirus, Bluetongue virus, Rotavirus A, Colorado tick fever virus, Aquareovirus A, Cypovirus 1, Fiji disease virus, Rice dwarf virus, Rice ragged stunt virus, Infectious pancreatic necrosis virus, Infectious bursal disease virus, Drosophila X virus, Saccharomyces cerevisiae virus L-A, Giardia lamblia virus, Leishmania RNA virus 1-1, Penicillium chrysogenum virus, Atkinsonella hypoxylon virus, White clover cryptic virus 1, White clover cryptic virus 2, Cryphonectria hypovirus 1-EP713, and Lettuce big-vein associated virus.

[0205] In certain embodiments, the viral RNA encodes genomic viral RNA of a virus from the order. In certain embodiments, the virus is a member of a fainily or subfamily that is selected from the group consisting of Paramyxoviridae, Pneumovirinae, Rhabdoviridae, Filoviridae, Bornaviridae, Orthomyxoviridae, Bunyaviridae, and Arenaviridae.

[0206] In certain embodiments, the virus is a inember of a genus that is selected from the group consisting of Respirovirus, Morbillivirus, Rubulavirus, Henipavirus, Avulavirus, Pneumovirus, Metapneumovirus, Vesiculovirus, Lyssavirus, Ephemerovirus, Cytorhabdovirus, Nucleorhabdovirus, Novirhabdovirus, Marburgvirus, Ebolavirus, Bornavirus, Influenzavirus A, Influenzavirus B, Influenzavirus C, Thogotovirus, Isavirus, Orthobunyavirus, Hantavirus, Nairovirus, Phlebovirus, Tospovirus, Arenavirus, Ophiovirus, Tenuivirus, or Deltavirus.

[0207] In certain embodiments, the virus is selected from the group consisting of Sendai virus, Measles virus, Mumps virus, Hendra virus, Newcastle disease virus, Human respiratory syncytial virus, Avian pneumovirus, Vesicular stomatitis Indiana virus, Rabies virus, Bovine ephemeral fever virus, Lettuce necrotic yellows virus, Potato yellow dwarf virus, Infectious hematopoietic necrosis virus, Lake Victoria marburgvirus, Zaire ebolavirus, Boma disease virus, Influenza A virus, Influenza B virus, Influenza C virus, Thogoto virus, Infectious salmon anemia virus, Bunyamwera virus, Hantaan virus, Dugbe virus, Rift Valley fever virus, Tomato spotted wilt virus, Lymphocytic chorioineningitis virus, Citrus psorosis virus, Rice stripe virus, and Hepatitis delta virus. In a specific embodiment, the virus is an Influenza A virus or an Influenza B virus. In another specific embodiment, the virus is an attenuated influenza virus, a cold adapted influenza virus, a temperature sensitive influenza virus, or a virus with any combination of these desirable properties. In one embodiment, the influenza virus incorporates an influenza B/Ann Arbor/l/66 strain virus, e.g., a cold adapted, temperature sensitive, attenuated strain of B/Ann Arbor/1/66. In another einbodiment, the influenza virus incorporates an influenza A/Ann Arbor/6/60 strain virus, e.g., a cold adapted, temperature sensitive, attenuated strain of A/Ann Arbor/6/60.

[0208] In certain embodiinents, the virus is a member of a family or subfamily that is selected from the group consisting of Leviviridae, Dicistroviridae, Picomaviridae, Sequiviridae, Comoviridae, Potyviridae, Caliciviridae, Astroviridae, Nodaviridae, Tetraviridae, Tombusviridae, Coronaviridae, Arteriviridae, Roniviridae, Togaviridae, Flaviviridae, Bromoviridae, Closteroviridae, Bamaviridae, Luteoviridae, Tymoviridae, and Flexiviridae.

[0209] In certain embodiments, the virus is a meinber of a genus that is selected from the group consisting of Levivirus, Allolevivirus, Cripavirus, Iflavirus, Enterovirus, Rhinovirus, Hepatovirus, Cardiovirus, Aphthovirus, Parechovirus, Sequivirus, Waikavirus, Comovirus, Fabavirus, Nepovirus, Potyvirus, Rymovirus, Bymovirus, Macluravirus, Ipomovirus, Tritimovirus, Vesivirus, Lagovirus, Norovirus, Sapovirus, Hepevirus, Mamastrovirus, Avastrovirus, Alphanodavirus, Betanodavirus, Betatetravirus, Omegatetravirus, Tombusvirus, Carmovirus, Necrovirus, Dianthovirus, Machlomovirus, Avenavirus, Aureusvirus, Panicovirus, Coronavirus, Torovirus, Arterivirus, Okavirus, Alphavirus, Rubivirus, Flavivirus, Pestivirus, Hepacivirus, Alfamovirus, Ilarvirus, Broinovirus, Cucumovirus, Oleavirus, Closterovirus, Crinivirus, Ampelovirus, Barnavirus, Luteovirus, Polerovirus, Enamovirus, Tobamovirus, Tobravirus, Hordeivirus, Furovirus, Pomovirus, Pecluvirus, Benyvirus, Idaeovirus, Sobemovirus, Umbravirus, Tymovirus, Marafivirus, Maculavirus, Aliexivirus, Manadrivirus, Carlavirus, Capillovirus, Foveavirus, Potexvirus, Trichovirus, Vitivirus, and Ourmiavirus.

[0210] In certain embodiments, the virus is selected from the group consisting of Enterobacteria phage MS2, Enterobacteria phage Q(3, Cricket paralysis virus, Infectious flacherie virus, Poliovirus, Human rhinovirus A, Hepatitis A virus, Encephalomyocarditis viius, Foot-and-mouth disease virus, Human parechovirus, Parsnip yellow fleck virus, Rice tungro spherical virus, Cowpea mosaic virus, Broad bean wilt virus 1, Tobacco ringspot virus, Potato virus Y, Ryegrass mosaic virus, Barley yellow mosaic virus, Maclura mosaic virus, Sweet potato mild mottle virus, Wheat streak mosaic virus, Swine vesicular exanthema virus, Rabbit hemorrhagic disease virus, Norwalk virus, Sapporo virus, Hepatitis E virus, Human astrovirus, Turkey astrovirus, Nodamura virus, Striped jack nervous necrosis virus, Nudaurelia capensis (3 virus, Nudaurelia capensis co virus, Tomato bushy stunt virus, Carnation mottle virus, Tobacco necrosis virus A, Carnation ringspot virus, Maize chlorotic mottle virus, Oat chlorotic stunt virus, Pothos latent virus, Panicum mosaic virus, Infectious bronchitis virus, Equine torovirus, Equine arteritis virus, Gill-associated virus, Sindbis virus, Rubella virus, Yellow fever virus, Bovine viral diarrhea virus, Hepatitis C
virus, Alfalfa mosaic virus, Tobacco streak virus, Brome mosaic virus, Cucumber mosaic virus, Olive latent virus 2, Beet ellows virus, Lettuce infectious yellows virus, Grapevine leafroll-associated virus 3, Mushrooin bacilliform virus, Barley yellow dwarf virus-PAV, Potato leafroll virus, Pea enation mosaic virus-1, Tobacco mosaic virus, Tobacco rattle virus, Barley stripe mosaic virus, Soil-borne wheat mosaic virus, Potato mop-top virus, Peanut clump virus, Beet necrotic yellow vein virus, Raspberry bushy dwarf virus, Southern bean mosaic virus, Carrot mottle virus, Turnip yellow mosaic virus, Maize rayado fino virus, Grapevine fleck virus, Shallot virus X, Indian citrus ringspot virus, Carnation latent virus, Apple stein grooving virus, Apple stem pitting virus, Potato virus X, Apple chlorotic leaf spot virus, Grape vine virus A, and Ourmia melon virus.

[0211] In certain einbodiments, the virus is a member of the family Namaviridae.

[0212] In certain einbodiments, the virus is a member of a genus that is selected from the group consisting of Narnavirus and Mitovirus.

[0213] In certain embodiments, the virus is selected from the group consisting of [0214] In certain embodiments, the virus is a member of a family or subfamily that is selected from the group consisting of Saccharomyces cerevisiae 20SRNA
nariiavirus and Cryphonectria parasitica mitovirus- 1 NB63 1.

6. Specific Embodiments [0215] 1. A host cell comprising a nucleic acid encoding a protease or pro-protease, wherein the nucleic acid encoding the protease or pro-protease is integrated into the host cell's genome, and wherein the protease or pro-protease is not normally expressed in the cell.

[0216] 2. The host cell of embodiment 1, wherein the cell expresses a protease.

[0217] 3. The host cell of embodiment 2, wherein the protease is a serine protease.
[0218] 4. The host cell of embodiinent 3, wherein the serine protease is an S
1 family protease.

[0219] 5. The host cell of einbodiment 4, wherein the protease is trypsin.

[0220] 6. The host cell of embodiment 2, wherein the serine protease is a bacterial subtilisin.

[0221] 7. The host cell of embodiment 6, wherein the protease is SPRT from Stfrepto3nyCes griseus.

[0222] 8. The host cell of embodiment 2, wherein the protease is a protease listed in Table 1.

[0223] 9. The host cell of embodiment 1, wherein the protease is a pro-protease.
[0224] 10. The host cell of embodiment 9, wherein the pro-protease is trypsinogen or prepro-SPRT protease from Streptoinyces griseus.

[0225] 11. The host cell of any of the preceding einbodiments, wherein expression of the protease or pro-protease is under the control of an inducible promoter.

[0226] 12. The host cell of embodiment 11, wherein the inducible promoter is induced by interferon or a downstreain signaling molecule induced by interferon.

[0227] 13. The host cell of einbodiment 11, wherein the inducible promoter is induced by a tetracycline-regulated expression system.

[0228] 14. The host cell of any one of embodiments 1-10, wherein expression of the protease or pro-protease is under the control of a constitutively active promoter.

[0229] 15. The host cell of any of the preceding embodiments, wherein the nucleic acid comprises a secretion signal that directs secretion of the protease or pro-protease.

[0230] 16. The host cell of any of the preceding einbodiments, wherein the cell is a mammalian cell.

[0231] 17. The host cell of embodiment 16, wherein the cell is a canine cell.
[0232] 18. The host cell of embodiment 17, wherein the cell is an MDCK cell.
[0233] 19. The host cell of embodiment 16, wherein the cell is a priinate cell.

[0234] 20. The host cell of embodiment 19, wherein the cell is an African green monkey or huinan cell.

[0235] 21. The host cell of any of embodiments 1-15, wherein the cell is an avian cell.
[02361 22. The host cell of embodiment 21, wherein the cell is a chicken cell.

[0237] 23. The host cell of any of the preceding embodiments, wherein the cell grows in suspension without being adapted.

[0238] 24. A method of producing an influenza virus comprising infecting the host cell of any of the preceding embodiments with an influenza virus, culturing the cell under conditions that allow replication of the influenza virus, and collecting influenza virus from the cell culture.

[0239] 25. A method of producing an influenza virus, comprising transfecting the host cell of any one of embodiments 1 to 23 with nucleic acids encoding an influenza genome, culturing the cell under conditions that allow replication of the influenza virus, and collecting influenza virus from the cell culture.

[0240] 26. The method of embodiment 24 or 25, wherein the host cell expresses a pro-protease, and wherein the method further coinprises adding an exogenous protease to the culture medium.

[0241] 27. The method of embodiment 26, wherein the exogenous protease is added to a maximum concentration of about 0.1 gg/hnl.

[0242] 28. The method of embodiment 26, wherein the exogenous protease is trypsin.

[0243] 29. A method of replicating an influenza virus in the absence of exogenously added trypsin comprising infecting a host cell with an influenza virus, culturing the cell under conditions that allow replication of the influenza virus, wherein said conditions do not include exogenously added trypsin, and collecting influenza virus from the cell culture.

[0244] 30. A method of replicating an influenza virus in the absence of exogenously added trypsin comprising transfecting a host cell with nucleic acids encoding an influenza genome, culturing the cell under conditions that allow replication of the influenza virus, wherein said conditions do not include exogenously added trypsin, and collecting influenza virus from the cell culture.

[0245] 31. The method of embodiment 29 or 30, wherein the host cell expresses an enzymatically active protease or pro-protease not norinally expressed by the cell.

[0246] 32. The method of embodiment 29 or 30, wherein the host cell is a mammalian cell.
[0247] 33. The method of enzbodiment 29 or 30, wherein the host cell is an avian cell.
[0248] 34. The method of embodiment 32, wherein the host cell is a primate cell, canine cell, hamster cell, mouse cell, or rat cell.

[0249] 35. The method of embodiment 32, wherein the host cell is an MDCK cell.
[0250] 36. The method of embodiment 34, wherein the host cell is a Vero cell.
[02511 37. The method of embodiment 33, wherein the host cell is a chicken cell.
[0252] 38. A method of increasing the titer of influenza virus grown in cell culture, comprising culturing an influenza virus in a cell culture, wherein the cells in the cell culture stably expresses a protease or pro-protease that i) is heterologous to the cell, and ii) cleaves a heinagglutinin of the influenza virus, thereby increasing the titer of the influenza virus grown in the cell culture relative to the titer obtained by culturing the influenza virus in cells that do not express a heterologous protease or pro-protease.

[0253] 39. The method of embodiment 38, wherein the cells constitutively express the protease or pro-protease.

[0254] 40. The method of embodiinent 38, wherein the cells inducibly express the protease or pro-protease.

[0255] 41. The method of embodiment 38, 39 or 40, wherein the cell expresses a protease.

[0256] 42. The method of embodiment 38, 39 or 40, wherein the cell expresses a pro-protease.

[0257] 43. The method of embodiment 41 wherein the protease is trypsin.

[0258] 44. The method of embodiment 41, wherein the protease is SPRT protease froin Streptoinyces gf=iseus.

[0259] 45. The method of embodiment 42, wherein the pro-protease is trypsinogen or prepro-SPRT protease from Streptonzyces griseus.

[0260] 46. A method for producing a heterologous protease or pro-protease in a host cell capable of supporting influenza replication, comprising culturing a cell comprising a nucleic acid encoding a protease or pro-protease not normally expressed in the cell under conditions that permit expression of said protease or pro-protease, thereby producing the protease or pro-protease in the cell.

[0261] 47. The method of embodiment 46, wherein the host cell stably expresses a protease.
[0262] 48. The inethod of embodiment 46, wherein the ho'st cell stably expresses a pro-protease.

[0263] 49. The method of embodiment 46, 47 or 48, wherein the host cell secretes the protease or pro-protease into the cell culture medium.

[0264] 50. The method of embodiment 46, 47, 48 or 49, wherein the host cell expresses between about 0.1 ng and about 10 g of the protease or pro-protease per ml of host cell culture.

[0265] 51. The method of embodiment 46, 47, 48, 49 or 50, wherein the cell expresses an amount of protease or pro-protease sufficient to increase the titer of virus grown in a culture of the host cells expressing the protease or pro-protease.

[0266] 52. The method of einbodiment 46, 47, 48, 49, 50 or 51, wherein expression of the protease or pro-protease is inducible.

[0267] 53. The method of embodiment 46, 47, 48, 49, 50 or 51, wherein expression of the protease or pro-protease is constitutive.

[0268] 54. An isolated nucleic acid encoding a nucleotide sequence that is at least about 90% identical to SEQ ID NO.:1.

[0269] 55. The nucleic acid of embodiment 54, wherein the nucleic acid encodes the nucleotide sequence of SEQ ID NO.:1.

[0270] 56. An isolated polypeptide comprising the amino acid of SEQ ID NO.:2.
[0271] 57. An isolated nucleic acid encoding the polypeptide of embodiinent 56.

[0272] 58. An expression vector comprising the nucleic acid of embodiment 54, 55 or 57.
[0273] 59. An isolated cell comprising the expression vector of einbodiment 58.

[0274] 60. A method of inaking a host cell that stably expresses a protease or pro-protease, comprising introducing a nucleic acid encoding a protease or pro-protease into a host cell, wherein the protease or pro-protease is not normally expressed in the host cell, and isolating the host cell that stably expresses the protease or pro-protease.

[0275] 61. A method of growing an adherent host cell in suspension comprising introducing a nucleic acid encoding a protease or pro-protease, wherein the nucleic acid encoding the protease or pro-protease is integrated into the host cell's genome, and wherein the protease or pro-protease is not normally expressed in the cell.

[0276] 62. The method of embodiment 60 or 61, wherein the host cell stably expresses a protease.

[0277] 63. The method of embodiment 60 or 61, wherein the host cell stably expresses a pro-protease.

[0278] 64. The method of embodiment 60, 61, 62 or 63, wherein the host cell secretes the protease or pro-protease into the cell culture medium.

[0279] 65. The method of embodiment 60, 61, 62, 63 or 64, wherein the host cell expresses between about 0.1 ng and about 50 g of the protease or pro-protease per ml of host cell culture.

[0280] 66. The method of embodiment 60, 61, 62, 63, 64 or 65, wherein the cell expresses an amount of protease or pro-protease sufficient to increase the titer of virus grown in a culture of the host cells expressing the protease or pro-protease.

[0281] 67. The method of embodiment 60, 61, 62, 63, 64, 65 or 66, wherein expression of the protease or pro-protease is inducible.

[0282] 68. The method of embodiment 60, 61, 62, 63, 64, 65 or 66, wherein expression of the protease or pro-protease is constitutive.

[0283] 69. The method of embodiment 60, 61, 62, 63, 64, 65, 66, 67 or 68, wherein the host cell is a mammalian cell.

[0284] 70. The method of einbodiment 60, 61, 62, 63, 64, 65, 66, 67 or 68, wherein the host cell is an avian cell.

[0285] 71. The method of embodiment 69, wherein the host cell is a primate cell, canine cell, hamster cell, mouse cell, or rat cell.

[0286] 72. The method of embodiment 71, wherein the host cell is an MDCK cell.
[0287] 73. The method of embodiment 71, wherein the host cell is a Vero [0288] 74. The method of embodiment 70, wherein the host cell is a chicken cell.
[0289] 75. The method of embodiment 60, wherein the host cell is a bacterial cell.

[0290] 76. The method of any one of embodiments 60-62 or 64-75, wherein the protease is trypsin.

[0291] 77. The method of any one of embodiments 60-62 or 64-75, wherein the protease is SPRT protease from Streptomyces gf iseus.

[0292] 78. The method of any one of embodiments 60-61 or 63-75, wherein the pro-protease is trypsinogen.

7. Examples [0293] The following examples serve merely to illustrate the invention and are not intended to limit the invention in any way.

7.1 Example 1: Growth of Influenza Strains in MDCK Cells [0294] This example describes characterization of several cell lines for culturing influenza.
Several different cell lines and primary cells were evaluated for the production of genetic reassortants derived from two laboratory adapted influenza strains, type A and type B, including MRC-5, WI-38, FRhL-2, PerC6, 293, NIH 3T3, CEF, CEK, DF-1, Vero, and MDCK. While many of the cell types supported the replication of some cold-adapted influenza strains to a limited extent, only MDCK consistently produced high titers of both type A and type B viruses. The MDCK cells were also tested for their ability to support replication of a potential pandemic vaccine, ca A/Vietnam/1203/2004. MDCK
cells were infected at a low multiplicity of infection with ca A/Vietnam/1203/2004 and virus in the supematant was quantified at various times post infection. By 48 hours post infection, the titers of ca A/Vietnam/1203/2004 reached approximately 8 loglo TCID50/mL and remained stable for the next 3 to 4 days. See Figure 1.

[0295] In the experiments, MDCK cells obtained from the ATCC (Accession No.
CCL-34) were expanded a limited number of times in either media containing 10% fetal bovine serum sourced from the United States or in an appropriate seruin free media (e.g., SFMV 100) to produce pre-master cell stocks for initial characterization studies.
Appropriate serum free media are described in U.S. Provisional Application No. 60/638,166, filed December 23, 2004, and in U.S. Provisional Application No. 60/641,139, filed January 5, 2005. Cells were readily grown in both types of media and both stocks of cells supported the replication of cold-adapted vaccine strains and pandemic strains as shown in Table 2, below, and in Figure 1, respectively.

Table 2 Coinparison of productivity of cold-adapted influenza strains in serum and serum free grown MDCK cells.
TCIDso/mL (logio) Virus strain (6:2 reassortant) MDCK with serum MDCK w/out serum A/New Caledonia/20/99 (H1N1) 8.1 7.8 A/Panama/20/99 (H3N2) 6.8 6.4 A/S dne /05/97 (H3N2) 7.0 6.5 B/Brisbane/32/2002 7.2 7.5 B/Hong Kona/330/2001 7.2 7.4 B/Victoria/504/2000 6.9 7.5 7.2 Example 2: Tumorigenicity of MDCK Cell Lines [0296] The potential tumorigenicity of the two pre-master cell stocks of MDCK
cells, one grown in media containing serum and the other in serum free media, were evaluated in the athymic nude mouse model at a stage that would represent 5 cell passages after that expected to be used for vaccine production. To evaluate tumorigenicity, 107 cells were injected subcutaneously into groups of 10 mice and after 84 days the animals were sacrificed and examined. Neoplasias were observed in six of the 10 animals inoculated with the cells passaged in serum free media. In contrast, there was no evidence of neoplasia in any of the animals inoculated with cells passaged in inedia supplemented with 10% fetal bovine serum;
although some fibrosarcomas were observed at the site of inoculation, cells passaged in seruin were not tumorigenic as shown in Table 3.

Table 3 Tumorigenicity and Karyology of MDCK cells passed in two different media Serum free 10% Serum Passage 4 Passage 20 Passage 4 Passage 20 Neoplasias No neoplasias.
Tumorigenicity ND noted ND Fibrosarcomas at injection site Estimated TPsoy 7 (no animals with ~'10 Not estimable tumors / total ND ND (>1()7) animals) (6/10) (0/10) 78; Large 78; Large 78; Few cells distribution of distribution of with 78; Few cells Karyology cells with cells with anomalous with anomalous Median number; chromosome comments chromosome chromosome chromosome number (70 to number of52 nuinber of 52- number (70 to 82) to 82 82 82) *TP50: Number of cells required to induce tuinors in 50% of animals ND: Not done [0297] As shown in Table 3, karyotype analyses were also performed on these two premaster cell stocks at both the fourth and twentieth passage in their respective media. The non-tumorigenic cells passaged in 10% FCS had a median number of 78 metaphase chromosomes with relatively limited distribution of cells with other chromosome numbers (70 to 82).
While the cells passaged in serum free media also had a median nuinber of 78 metaphase chromosomes, significantly more cells were observed witll an aneuoploid chromosome number ranging from 52 to 82 metaphase chromosomes. In both cases, the karyology did not change following passage.

7.3 Example 3: Adapting MDCK Cells to Grow in Serum Free Media [0298] MDCK cells from the ATCC are passaged in media containing gamma irradiated FBS. These cells are then passaged a limited number of times in a serum free media formulation chosen to support cell bank production. Seruin free media are described in U.S.
Provisional Application Nos. 60/638,166 and 60/641,139. These additional passages may be performed at either 37 C or 33 C. Passage of MDCK cells in three media containing plant-derived supplements rather than serum yielded cells with karyotypes similar to that of MDCK
cells passaged in FCS containing media (data not shown).

7.4 Example 4: Cloning of MDCK Cells [0299] Cells were biologically cloned through limiting dilution in order to ensure that the production cells are derived from a unique genetic constellation. Clones were screened for various phenotypic properties including doubling time and relative tuinorigenicity, as well as viral production. In an initial proof of concept experiment, fifty-four MDCK
clones were obtained in media containing FCS. These clones were passaged and each was infected with a low multiplicity of infection of ca A/New Caledonia/20/99. Several days after infection, the supematant was removed and the quantity of virus in the supernatant was measured by TCID50. A minority of the clones produced relatively high titers of virus, greater than was produced in the noncloned parental cells. Clones with superior biological and physiological properties are used to establish a Master Cell Bank (MCB) as described below.

7.5 Example 5: Testing and Characterization of a Master Cell Bank [0300] The MCB is extensively tested to ensure that there is no evidence of adventitious agents. For example, one or more of several PCR and/or antibody-specific tests for available viral agents are conducted, as shown in Table 4, below.

Table 4 Testing regimen for the MCB
General tests PCR* / Ab specific Sterility AAV Types 1 &2 Mycoplasma HCMV
Adventitious agents in vitro (multiple cell lines) EBV
Adventitious agents in vivo HSV
PERT Hepatitis B, C & E
Co-cultivation HHV 6, 7& 8 Karyology HIV 1 &2 Electron microscopy HPV
Tumorigenicity intact cells (TP50) HTLV I & II
Oncogenicity of cellular DNA Polyoma (BK and JC viruses) Oncogenicity of cellular lysate Circovirus Bovine viruses per 9CFR Canine Parvovirus Porcine viruses per 9CFR Canine distein er Adenovirus 7.6 Example 6: Preclinical Characterization of Cell Culture-Derived Influenza Virus [0301] This exainple describes characterization of influenza strains produced fiom cell culture as well as from eggs and compares the viruses produced from the systems. Generally, the influenza viruses are suitable for use as vaccines in huinans, and have biological properties that make the viruses suitable for such use. In this exainple, the influenza viruses are cold-adapted (ca; have the ability to replicate efficiently at lower temperatures), temperature sensitive (ts; have restricted replication in vitro at higher temperatures), and attenuated (att; no detectable replication in lung tissues of ferrets), and are referred to herein as ca ts att strains. The comparison includes: biochemical, antigenic, and genetic evaluation (sequencing) of viral product; biological and biocheinical characterization of the virus following replication in human cells; replication in a permissive animal model; and immunogencity in a permissive animal model.

7.6.1 Genetic, biochemical and antigenic comparability [0302] Ca ts att strains of type A/H1N1, A/H5N1, A/H3N2 and B replicated to relatively high titers in MDCK cells. In addition, passaging these ca ts att strains in MDCK cells did not alter their genomic sequence. Three ca ts att strains, ca A/Sydney/05/97, ca A/Beijing/262/95, and ca B/Ann Arbor/l/94 were passaged once or twice in MDCK
cells and the entire coding regions of all 6 internal genes were sequenced and compared to the starting material. No nucleotide changes were observed, demonstrating that this passaging through this substrate did not change the genetic composition of these strains.
Further sequence characterizations is performed on different vaccine strains produced in MDCK
cells under conditions that are expected to mimic the production process including media composition, input dose (moi), temperature of incubation and time of harvest. Based on the preliminary data, it is expected that there will be no changes in the genomic sequence of MDCK-produced virus.

[0303] Because the genome was genetically stable following passage in MDCK
cell, the biological traits of the vaccine produced in eggs or MDCK cells are expected to be indistinguishable. However, the primary viral product from cell culture may have some subtle differences coinpared to the egg based product, particularly with respect to post-translational modification of viral proteins including HA and NA, or composition of lipids in the viral membrane; both of which could potentially change the overall physical properties of the virion. Preliminary preclinical data on the antigenicity of cell culture produced and egg produced vaccine deinonstrated that there were no detectable differences in this important parameter. Egg stocks of several vaccine strains were passaged through MDCK
cells and the antigenicity of both products was determined by measuring the HAI titers using reference antisera. As show in Table 5, all the HAI titers were within 2-fold of one another, indicating that replication of the vaccine in cells did not change the antigenicity of the vaccine compared to egg derived material.

Table 5 HAI Titers of strains produced in eggs and MDCK cells HAI Titer Strain Egg derived MDCK
derived A/Panama/20/99 256 256 A/Wuhan/359/95 1024 2048 A/W oniin /03/2003 512 1024 B/Jilin/20/2003 64 32 B/Hong Kong/330/01 64 64 B/Jiangsu/10/2003 128 128 7.7 Example 7: Infection of Human Epithelial Cells in Culture [0304] In certain embodiments, to evaluate the biochemical, biological, and structural similarities following replication of the MDCK and egg produced vaccines in cells of human origin, vaccines may be passaged once in relevant diploid huinan cells, such as norinal human bronchial epithelial cells (NHBE). This passage will serve to mimic a single infection event in the human airway and then enable comparison of the progeny virus, the virus that is ultimately responsible for eliciting an effective immune response. Studies of the vaccines' hemagglutinin (binding and fusion) and neuraminidase activities are measured on these materials as well as other biochemical and structural studies including electron microscopy, infectious to total particle ratios, and viral genome equivalents can be evaluated. Overall, these comparisons serve to demonstrate the comparability of the cell-derived vaccine to the effective and safe egg produced vaccine. A suminary of analytical studies is suinmarized in Table 6.

Table 6 Preclinical studies to compare cell and egg produced vaccines In vivo (ferrets) In vitro*
Attenuation / Replication Virus binding Extent of replication in upper airway Hemagglutination titer Kinetics of replication in upper airway Binding of different sialic acids Iminunogenicity Physical properties Cross-reactivity Morphology by EM
Kinetics Infectious : Total particles (genomes) Infectivity Fusion activity Dose required for detectable replication pH optimum Dose required for antibody response temperature optimum Genomic sequence Neuraininidase activity *Compare primary products and after one passage in human cells 7.8 Example 8: Preclinical Animal Models [0305] The ferret is a robust animal model used to evaluate the attenuation and immunogenicity of attenuated influenza vaccines and coinponent vaccine strains. The perfonnance of cell-derived influenza strains produced from the MCB are compared to the same strains produced in eggs. Head to head coinparison of these materials in controlled studies enables a high level of assurance of the comparability of these viral products.

[0306] In order to evaluate the ability of the two vaccines to infect or achieve a "take" in the ferret, animals are lightly anesthetized and inoculated intranasally with either the cell or egg produced viral preparations. Nasal wash material is collected at several time points following inoculation and the quantity of virus is evaluated by one of several available methods in order to evaluate the kinetics and extent of viral replication in the animals' upper respiratory tract.
Experiments are performed with a range of doses and include multiple strains and different trivalent mixtures to generalize the relative infectivity of cell culture grown strains to egg produced strains. These same studies are also used to evaluate the immunogenicity of the influenza strains, a property that is inherently linked to the ability of the virus to initiate infection. Aliimals are bled and nasal washes are harvested at various points (weeks) post inoculation; these specimens are used to assess the serum antibody and nasal IgA responses to infection. The culmination of these data, infectivity, seruin antibody and inucosal antibody responses, will be used to compare and evaluate the relative infectivity of the cell-produced vaccine to the egg produced vaccine. The most likely outcome is predicted to be that the cell and egg produced vaccine strains have similar infectivity and immunogenicity.
If the cell derived vaccine appeared to be more infective or more iminunogenic than the egg-derived product, further studies evaluating the possibility of lower dosage are performed.
[0307] A number of immunogenicity and replication studies are perforined in the ferret model to evaluate the cell culture-derived vaccines with a single unit human dose. Infection with catsatt strains generally elicits strong and rapid antibody responses in ferrets. In addition, individual catsatt strains are routinely tested and shown to express the attenuated (att) phenotype by replicating to relatively high titers in the nasopharynx but to undetectable levels in the lung of these animals. The impact of cell culture growth on these biological traits is also assessed. However, it is unlikely that any differences will be seen, since the att phenotype is an integral part of the genetic composition of these strains. The growth kinetics and crossreactivity of these strains is evaluated following administration of a single human dose in these animals. The selicits serum antibodies that cross-react with inultiple strains within a genetic lineage; and it is expected that a cell-derived vaccine will have the same capability.

[0308] These comparability evaluations should provide significant insight into potential biochemical and/or biophysical differences of the primary virus product and demonstrate the impact of these epigenetic differences on the performance of the catsatt strains measured by first passaging the virus in human cells or animal studies. Based on the sequence information to date, there is no expected impact on the catsatt strains iminunogenic performance resulting from production on MDCK cells.

[0309] Ferrets are a well docuinent animal model for influenza and are used routinely to evaluate the attenuation phenotype and immunogenicity of catsatt strains. In general, 8 - 10 week old animals are used to assess attenuation; typically study designs evaluate n=3-5 animals per test or control group. Immunogenicity studies are evaluated in animals fiom 8 weeks to 6 months of age and generally require n=3-5 animals per test article or control group. These numbers provide sufficient information to obtain statistically valid or observationally important comparisons between groups. During most studies Influenza-like signs may be noticed, but are not likely. Ferrets do not display signs of decrease in appetite or weight, nasal or ocular discharge; observing signs of influenza-like illness is a necessary part of the study and interventions such as analgesics are not warranted.
Other signs of discomfort, such as open sores or significant weight loss, would result in appropriate disposition of the animal following discussion with the attending veterinarian.

7.9 Example 9: Master virus seed (MVS) development [0310] Currently influenza vaccine strains are generated by co-infecting avian cells with a wild type virus and either the type A or type B MDV and isolating and screening the progeny for the desired 6:2 genetic constellation. This process requires several passages of the virus through avian cell cultures and/or SPF eggs. Recently, plasmid rescue has been introduced for producing influenza viral preparation. In this process, Vero (African green monkey) cells from an extensively tested and characterized cell bank are electroporated with plasmids, each containing a eDNA copy of one of the 8 influenza RNA segments.
Several days after electroporation the supernatant of these electroporated cells contains influenza virus. The supematants are then inoculated into SPF eggs to amplify and biologically clone the vaccine strain. Both of these procedures result in a vaccine strain that is inoculated into SPF eggs to produce the MVS. While plasmid rescue has several advantages including more reliable timing, more genetically accurate gene segments and less potential containination with adventitious agents from the wild type isolate, individual MVS's generated by these two methods are indistinguishable from one another and can be used to initiate bulk vaccine production.

[0311] Final anlplification of the vaccine strains is conducted in cells derived from the MDCK cell banks. This final amplification can be achievable with small-scale cultures (<20L) of MDCK cells. The supematant from these cells is collected, concentrated and characterized/tested to produce the MVS.

7.10 Example 10: Proteolytic Activation of Infectious Virus with MDCK Cells Expressing a Heterologous Pro-Protease [0312] The following example describes construction of MDCK cell lines constitutively expressing porcine trypsinogen.

[0313] First, the gene encoding porcine trypsinogen was cloned into the retroviral vector pLNHX (Clonetech Inc., Mountain View, CA). To do so, a recombinant nucleotide sequence encoding trypsinogen, the sequence of which is presented as Figure 11 (SEQ ID
NO.:3) was synthesized and cloned into a shuttle vector. The trypsinogen gene was then digested out of the shuttle vector with Bgl 11 and cloned into the Bgl II site of the pLNHX
polylinker. The vector thus produced is referred to herein as pTGEN.

[0314] Next, pTGEN was transfected into two different packaging cell lines, Ampho 293 and GP2, using conventional techniques. Retrovirus containing pTGEN (referred to herein as vTGEN) or a luciferase expression vector (referred to herein as vLLRN) for use as a positive control were isolated 48 hours post-infection by lysing the packaging cells using conventional techniques. Infection of MDCK cells with vLLRN indicated that vLLRN produced in GP2 cells could 1) introduce the luciferasae gene into the MDCK genome, as luciferase activity could be detected in MDCK cells at 48 hours post infection (Figure 2) and 2) the amount of luciferase expressed by the MDCK cells at 48 hours post infection was proportionate to the concentration of vLLRN used to infect the MDCK cells (Figure 3). Thus, the positive controls demonstrate that the pLNHX systein can be used to transfect MDCK
cells with heterologous genes, and that such genes are expressed by the MDCK cells.

[0315] To identify clones stably transfected with pTGEN, the following procedure was used.
48 hours post-infection with vTGEN, vLLRN, or vLNHX (vector-only negative control), G418 was added to the growth medium to select for resistant MDCK clones.
Approximately 3 weeks later, individual MDCK clones were isolated and expanded as isogenic cultures.
[0316] Expression of luciferase from stably-transformed MDCK clones is shown in Figure 4.
As seen in Figure 4, 12 different individual clones and two different mixtures of 3 clones expressing luciferase at high levels were obtained by this procedure. The 12 individual clones produced between about 10 ng to 300 ng active luciferase per 1000 cells. Thus, these data deinonstrate that stably-transformed MDCK clones expressing trans-genes at high levels can be obtained from pLNHX vector systems.

[0317] A suininary of the individual clones and clone mixes obtained from the transfection experiments is presented as Figure 5. As shown in Figure 5, 18 MDCK clones expressing trypsinogen each were isolated fiom retroviral particles produced in Ampho 293 cells and in GP2 cells.

7.11 Example 11: Influenza Infection and Growth in MDCK Cells Expressing Trypsinogen in Culture [0318] This example describes infection and growth of two different influenza strains in MDCK cells that express trypsinogen in the presence and absence of exogenous trypsin.
[0319] The ability of two MDCK clones expressing trypsinogen (one produced by transfecting MDCK cells with particles produced in Ampho 293 cells, the other with particles in produced in GP2 cells) to serve as a viral host was assessed as follows.
Cells from the appropriate clone were infected with either strain ca A/Ann Arbor/6/60 MDV-A
or ca A/New Caladonia/20/90 at a multiplicity of infection of 0.01 (i.e., 1 influenza virus per 100 cells) at time 0. Cells transfected with pLLRN or pLNHX were used as controls. Viral titer in the cultures was assessed at 12 hours, 24, hours, 48 hours, 72 hours, 96 hours, and 120 hours following infection.

[0320] In one group of experiments, exogenous porcine trypsin was added to the cell culture to a final concentration of 0.1 gg/ml at 24 hours following infection (closed circles). In another group of experiments, the concentration of exogenous porcine trypsin in the culture was maintained at 1 ghnl from day 1 (24 hours following infection) until day 5 (120 hours following infection). In the last group of experiments, no exogenous trypsin was added.

[0321] The results of the experiments are shown as Figures 6A-6C. Figure 6A
present the results of influenza infection of mock-infected MDCK cells with MDV-A (top left) or ca A/NC (top right) and of luciferase-expressing MDCK cells with MDV-A (bottom left) or ca A/NC (bottom right). As expected, overall viral yield was substantially greater for all experiments when exogenous trypsin was added to I g/ml from day 1 to day 5 (open squares) and yields were lowest when no exogenous trypsin was added (open triangles).
Addition of exogenous trypsin at 0.1 ghnl on day I did not significantly increase viral yield (closed circles).

[0322] Figures 6B-6C present the results of influenza infection of two different trypsinogen-expressing MDCK clones with MDV-A (left panels) or ca A/NC (right panels). The MDCK
clones used in the experiment shown in Figures 6B were made by transfection with viral particles produced in Ainpho 293 cells, while the MDCK clones used in these experiment shown in Figures 6C were made by transfection with viral particles produced in GP2 cells [0323] As shown in Figures 6B-6C, viral titers were significantly increased when the infected cells expressed trypsinogen. In all cases, viral titers obtained in the absence of exogenous trypsin (open triangles) were less than those obtained in the presence of 1 g/ml trypsin (open squares), though greater than the viral titers observed for the controls.
However, in several cases (e.g., Fig. 6B top left, top right and bottom right; Fig. 6C top left and bottom right), addition of a small amount of trypsin (0.1 g/ml) at day 1(closed circles) increased resulting viral titers to be comparable to those observed in the presence of exogenous trypsin. Without intending to be bound to any particular theory or mechanism of action, it is believed that the addition of exogenous trypsin was sufficient to cleave the pro-peptide of trypsinogen, thereby activating the proenzyrne to its fully active form. Thus, "priming" the culture with a small amount of peptidase can activate the proenzyme produced by the cells in the culture, increasing overall viral yields.

7.12 Example 12: Expression of Tagged Trypsinogen in MDCK Cells [0324] This exainple describes production and detection of trypsinogen tagged with six histidine residues in MDCK cells. 1 [0325] To identify clones expressing trypsinogen at high levels, another set of clones were made by transfecting a gene encoding a 6xHis-labeled trypsinogen fusion protein into MDCK
cells. Briefly, the recombinant nucleotide sequence encoding porcine trypsinogen presented as Figure 11 was cloned into the polylinker of pDESTTM26 (Invitrogen Inc., Carlsbad, CA) and the resulting vector transfected into MDCK cells. The pDESTTM26 plasmid encodes the 6x His tag adjacent to the polylinker, such that expression of the artificial trypsinogen gene from pDESTTM26 yields 6x His-tagged porcine trypsinogen. The transfected cells were then cultured for 48 hours, then transfectants were selected by culturing for about 3 weeks in the presence of G418. 16 G418-resistant clones were then isolated and expanded as single clones.

[0326] Expression of trypsinogen by the 16 clones was then assessed by western blot.
Briefly, each clone was cultured for 1-2 days, then protein was isolated from the cells by collecting the cells by centrifugation, washing the cells once with lx phosphate buffered saline, then lysing the cells in 200 l of 1x protein lysis buffer (Promega Corp.; Madison, WI). Next, 10 [t1 of each protein lysate was loaded on a 12% denaturing polyacrylimide gel for Western blot analysis. 6xHis-labeled trypsinogen was detected with a rabbit anti-trypsin polyclonal antibody (Chemicon AB 1823; Chemicon International; Temecula, CA) specific for trypsin diluted at 1:10,000. Goat anti-rabbit HRP conjugated secondary antibody diluted at 1:3,000 was added, washed twice, and protein visualized by addition of ECL
plus substrate (GE Healthcare Bio-Sciences Corp.; Piscataway, NJ). The results of the western blots are presented as Figures 7A and 7B. As shown in these figures, 12 of 16 G418 resistant MDCK
clones produced 6xHis-labeled trypsinogen in detectable amounts.

7.13 Example 13: Expression of Trypsin in MDCK Cells Under the Control of an Inducible Promoter [0327] This example describes the construction of MDCK cell lines that express trypsin under the control of an inducible promoter.

[0328] To test another way to provide trypsin to cell cultures at the appropriate time following viral infection, MDCK cells that express trypsin under the control of an inducible promoter were made as follows. First, MDCK cell lines were transfected with pcDNATM6/TR
(Invitrogen, Inc., Carlsbad, CA) and transfectants were selected by culturing in the presence of blasticidin. Antibiotic-resistant clones were then isolated and expanded from single clones.

[0329] Next, expression of the tetracycline (tet) repressor protein from the integrated vector in the individual clones was tested with the positive control vector pDEST30Luc.
(Invitrogen, Inc., Carlsbad, CA). The goal of this experiment was to identify MDCK clones that express sufficient tet repressor protein to have very low luciferase expression in the absence of doxycycline (dox) and high expression in the presence of dox. To do so, approximately 20,000 cells from each clone to be screened were placed in each well of a 6-well microtiter plate. Each sample was transiently transfected with pDEST30Luc using conventional techniques, then dox was added to 3 of the wells at one day following transfection. The cells were harvested at two days following transfection, and 20 l supematant was used for each luciferase assay.

[0330] Results of the luciferase assays for 12 clones are presented as Figure 8. As shown in Figure 8, clone 3 exhibited the greatest difference between activities observed in the presence and absence of doxycycline. 36 MDCK-Trex clones were screened with the transient Luciferase assay. Two clones MDCK/R3 and MDCIQR7 were selected for making inducible Trypsin expression MDCK cells (fold induction: +DC/-DC=13.1 and 14.0, respectively).
MDCK/R3 or R7 were transfected with pT-Rex-DEST30/Luciferase (Figure 13) as a control.
29 antibiotics resistant clones R3/Luc and R7/Luc were isolated and expanded for Luciferase assay with or without Doxycycline (DC) induction. There is up to a 70-fold difference for R3 and up to a 56-fold difference for R7 after the chemical induction (+DC/-DC=
70 and 56).
Our data indicate that R3/Luc and R7/Luc clones stably carried Luciferase gene and that Luciferase expression is inducible by DC.

[0331] To make inducible trypsin expression MDCK cells, MDCK/R3 or R7 were transfected with pT-Rex-DEST30/Trypsin (Figure 13) and antibiotics resistant clones were selected with MDCK growth medium+Blasticidin/G418 for 2-3 weeks. 70 clones expressing trypsin were isolated and expanded for protease activity assays. Genomic DNAs from these clones were isolated and trypsin gene was detected by PCR. No DNA nucleoside inutations were found by DNA sequence analysis.

[0332] Next, the clones exhibiting the greatest difference between activities observed in the presence and absence of doxycycline are evaluated by growth curve studies such as those described in Exainple 11 to assess growth and titers of influenza viruses cultured on cells inducibly expressing trypsinogen. Approximately 15 clones had high trypsin with DC
induction and low trypsin expression without DC. For example, one clone R3/6U7 has baseline Trypsin expression of -10 ng/ml in the absence of DC which increases to -2.5 g/ml (-250 fold induction) when induced with 3 g/ml DC. For the replication of most influenza vaccine strains trypsin is generally used at a concentration of -1.0 ghnl.
The expression level of trypsin in these cells is induced and controlled with different DC
concentrations for flu vaccine production.

[0333] Many inducible trypsin expression MDCK clones are perinissive for infection and replication of influenza strains useful for the production of vaccines. The parental MDCK
cells generally grow as adherent cells with less then 10% of the cells in suspension.

However; the inducible trypsin MDCK clones are less adherent with about 30% of the cells growing in suspension cells. The majority of the cells in suspension (>90%) are viable by trypan blue staining analysis. By adjusting the endogenous trypsin expression, the percent of cells in suspension may be increased. The use of suspension cells for flu vaccine production can reduce the cost of goods by eliininating the need for carriers.

7.14 Example 14: Cloning of a Bacterial Serine Protease from Streptofizyces griseus [0334] This example describes the cloning of a serine protease from the bacterium Stfreptonzyces griseus.

[0335] To do so, a priiner pair (Spr T forward and reverse) was used to ainplify the coding sequence of the SPRT protease from genomic DNA using conventional techniques.
The nucleotide sequences of the primer pair are presented as Figure 12. A
Streptomyces gt iseus strain deposited with the ATCC (Accession No. 23915) was used as the source for the genomic DNA. The sprT gene was then cloned into pDESTTM14 using conventional techniques, and the nucleotide sequence of the sprT gene determined using conventional techniques.

[0336] The nucleotide sequence of the sprT gene is presented as Figure 9.

[0337] While the foregoing invention has been described in some detail for purposes of clarity and understanding, it will be clear to one skilled in the art from a reading of this disclosure that various changes in form and detail can be made without departing fiom the true scope of the invention. For exainple, all the techniques and apparatus described above may be used in various combinations. All publications, patents, patent applications, or other documents cited in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent, patent application, or other document were individually indicated to be incorporated by reference for all puiposes.
In addition, U.S. Provisional Patent Application Nos.: U.S. 60/ 793,522, filed April 19, 2006;
U.S. 60/ 793,525, filed April 19, 2006; U.S. 60/702,006, filed July 22, 2005;
U.S.
60/699,556, filed July 15, 2005; U.S. 60/699,555, filed July 15, 2005; U.S.
60/692,965 filed June 21, 2005; and U.S. 60/692,978 filed June 21, 2005, are incorporated by reference in their entirety for all purposes.

DEMANDE OU BREVET VOLUMINEUX

LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter le Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS

THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME:

NOTE POUR LE TOME / VOLUME NOTE:

Claims (22)

1. A host cell comprising a nucleic acid encoding a protease or pro-protease, wherein the nucleic acid encoding the protease or pro-protease is integrated into the host cell's genome, and wherein the protease or pro-protease is not normally expressed in the cell.
2. The host cell of claim 1, wherein the cell expresses a protease.
3. The host cell of claim 2, wherein the protease is a serine protease.
4. The host cell of claim 1, wherein the protease is a pro-protease.
5. The host cell of claim 1, wherein expression of the protease or pro-protease is under the control of an inducible promoter.
6. The host cell of claim 1, wherein expression of the protease or pro-protease is under the control of a constitutively active promoter.
7. The host cell of claim 1, wherein the nucleic acid comprises a secretion signal that directs secretion of the protease or pro-protease.
8. The host cell of claim 1, wherein the cell is a mammalian cell.
9. The host cell of claim 8, wherein the mammalian cell is a human cell, a primate cell, a canine cell, hamster cell, mouse cell, or rat cell.
10. The host cell of claim 1, wherein the cell is an avian cell.
11. The host cell of claim 1, wherein the host cell grows in suspension without adaptation.
12. A method of producing an influenza virus, comprising:
a) introducing an influenza genome into the host cell of claim 1 cell by infecting the host cell with an influenza virus, or transfecting the host cell with nucleic acids encoding an influenza genome;
b) culturing the cell under conditions that allow replication of the influenza virus;
and c) collecting influenza virus from the cell culture.
13. The method of claim 12, wherein the host cell expresses a pro-protease, and wherein the method further comprises adding an exogenous protease to the culture medium.
14. The method of claim 13, wherein the exogenous protease is added to a maximum concentration of about 0.1 µg/ml.
15. A method of replicating an influenza virus in the absence of exogenously added protease, comprising:
a) introducing an influenza genome into the host cell of claim 1 by infecting the host cell with an influenza virus or transfecting the host cell with nucleic acids encoding an influenza genome;
b) culturing the cell under conditions that allow replication of the influenza virus, wherein said conditions do not include exogenously added protease; and c) collecting influenza virus from the cell culture.
16. A method of increasing the titer of influenza virus grown in cell culture, comprising culturing the influenza virus in a cell culture, wherein the cells in the cell culture stably expresses a protease or pro-protease that i) is heterologous to the cell, and ii) cleaves a hemagglutinin of the influenza virus, thereby increasing the titer of the influenza virus grown in the cell culture relative to the titer obtained by culturing the influenza virus in cells that do not express a heterologous protease or pro-protease.
17. A method for producing a heterologous protease or pro-protease in a host cell capable of supporting influenza replication, comprising culturing a cell comprising a nucleic acid encoding a protease or pro-protease not normally expressed in the cell under conditions that permit expression of said protease or pro-protease, thereby producing the protease or pro-protease in the cell.
18. The method of claim 17, wherein the cell expresses an amount of protease or pro-protease sufficient to increase the titer of virus grown in a culture of the host cells expressing the protease or pro-protease.
19. An isolated nucleic acid comprising SEQ ID NO.:1.
20. An isolated polypeptide comprising the amino acid of SEQ ID NO.:2.
21. An expression vector comprising the nucleic acid of claim 19.
22. An isolated cell coinprising the expression vector of claim 21.
CA002613283A 2005-06-21 2006-06-20 Methods and compositions for expressing a heterologous protease Abandoned CA2613283A1 (en)

Applications Claiming Priority (15)

Application Number Priority Date Filing Date Title
US69297805P 2005-06-21 2005-06-21
US69296505P 2005-06-21 2005-06-21
US60/692,965 2005-06-21
US60/692,978 2005-06-21
US69955505P 2005-07-15 2005-07-15
US69955605P 2005-07-15 2005-07-15
US60/699,556 2005-07-15
US60/699,555 2005-07-15
US70200605P 2005-07-22 2005-07-22
US60/702,006 2005-07-22
US79352506P 2006-04-19 2006-04-19
US79352206P 2006-04-19 2006-04-19
US60/793,522 2006-04-19
US60/793,525 2006-04-19
PCT/US2006/023866 WO2007002007A2 (en) 2005-06-21 2006-06-20 Methods and compositions for expressing a heterologous protease

Publications (1)

Publication Number Publication Date
CA2613283A1 true CA2613283A1 (en) 2007-01-04

Family

ID=37595721

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002613283A Abandoned CA2613283A1 (en) 2005-06-21 2006-06-20 Methods and compositions for expressing a heterologous protease
CA002613284A Abandoned CA2613284A1 (en) 2005-06-21 2006-06-20 Methods and compositions for expressing negative-sense viral rna in canine cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA002613284A Abandoned CA2613284A1 (en) 2005-06-21 2006-06-20 Methods and compositions for expressing negative-sense viral rna in canine cells

Country Status (11)

Country Link
US (4) US20060286591A1 (en)
EP (2) EP1917352A4 (en)
JP (2) JP2008543331A (en)
KR (2) KR20080024212A (en)
AU (2) AU2006262381A1 (en)
BR (1) BRPI0612268A2 (en)
CA (2) CA2613283A1 (en)
IL (1) IL188202A0 (en)
NO (1) NO20080358L (en)
RU (1) RU2008101367A (en)
WO (2) WO2007002007A2 (en)

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7763609B2 (en) 2003-12-15 2010-07-27 Schering Corporation Heterocyclic aspartyl protease inhibitors
US11865172B2 (en) 2005-04-21 2024-01-09 University Of Florida Research Foundation, Inc. Materials and methods for respiratory disease control in canines
US7959929B2 (en) 2005-04-21 2011-06-14 University Of Florida Research Foundation, Inc. Materials and methods for respiratory disease control in canines
EP1917352A4 (en) 2005-06-21 2009-03-25 Medimmune Vaccines Inc Methods and compositions for expressing a heterologous protease
US7790434B2 (en) 2005-06-21 2010-09-07 Medimmune, Llc Methods and compositions for expressing negative-sense viral RNA in canine cells
US7682619B2 (en) 2006-04-06 2010-03-23 Cornell Research Foundation, Inc. Canine influenza virus
DK2016194T3 (en) * 2006-04-19 2015-06-15 Medimmune Llc METHODS AND COMPOSITIONS FOR EXPRESSION OF VIRAL NEGATIVE SENSE RNA IN DOG CELLS
WO2008032219A2 (en) 2006-09-11 2008-03-20 Novartis Ag Making influenza virus vaccines without using eggs
US9163053B2 (en) * 2007-05-18 2015-10-20 Fluidigm Corporation Nucleotide analogs
JP5540329B2 (en) 2007-06-26 2014-07-02 公益財団法人ヒューマンサイエンス振興財団 Vaccine antigens that induce cross-neutralizing antibodies against high-risk group human papillomaviruses
GB0810305D0 (en) 2008-06-05 2008-07-09 Novartis Ag Influenza vaccination
US8785173B2 (en) 2008-09-24 2014-07-22 Medimmune, Llc Methods for purification of viruses
EP2424565A1 (en) 2009-04-27 2012-03-07 Novartis AG Adjuvanted vaccines for protecting against influenza
PL2401384T3 (en) * 2009-05-21 2013-03-29 Seqirus Uk Ltd Reverse genetics using non-endogenous pol i promoters
CN102666860B (en) 2009-07-31 2015-06-17 诺华股份有限公司 Reverse genetics systems
CN102220437B (en) * 2011-04-27 2014-04-23 贵州省烟草科学研究所 Method for identifying viral disease resistance of tobacco through partial inoculation
AU2013205478B9 (en) 2012-03-02 2014-11-20 Seqirus UK Limited Influenza virus reassortment
GB201220119D0 (en) * 2012-11-08 2012-12-26 Univ Cork Vector
JP6421128B2 (en) 2012-12-03 2018-11-07 ノバルティス アーゲー Rear sortant influenza A virus
BR112015021880A2 (en) 2013-03-13 2017-09-26 Novartis Ag influenza b virus rearrangement
DE202013005130U1 (en) 2013-06-05 2013-09-10 Novartis Ag Influenza virus reassortment
DE202013005100U1 (en) 2013-06-05 2013-08-26 Novartis Ag Influenza virus reassortment
JP2016521553A (en) 2013-06-06 2016-07-25 ノバルティス アーゲー Influenza virus reassembly
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
JP6748067B2 (en) * 2015-03-17 2020-08-26 一般財団法人阪大微生物病研究会 Influenza virus production cells
CN106520700B (en) * 2016-11-10 2019-12-27 中国食品药品检定研究院 MDCK cell line for stably expressing bovine trypsinogen and application thereof
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
JPWO2020004425A1 (en) * 2018-06-27 2021-07-08 一般財団法人阪大微生物病研究会 Influenza virus culture method
CN109762938B (en) * 2019-02-01 2022-06-10 中国水产科学研究院珠江水产研究所 Primer combination and kit for distinguishing mandarin fish rhabdovirus vRNA, cRNA and mRNA
CN109652384B (en) * 2019-02-21 2021-10-26 昆明理工大学 Method for culturing hepatitis E virus in vitro
BR102020010208A2 (en) 2020-05-21 2022-04-12 Fundação Oswaldo Cruz Nucleic acid construction, recombinant influenza virus, method of making a recombinant influenza virus, composition, and use.

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6001634A (en) * 1989-08-28 1999-12-14 Palese; Peter Recombinant negative strand RNA viruses
US6887699B1 (en) * 1990-05-22 2005-05-03 Medimmune Vaccines, Inc. Recombinant negative strand RNA virus expression systems and vaccines
US6348327B1 (en) * 1991-12-06 2002-02-19 Genentech, Inc. Non-endocrine animal host cells capable of expressing variant proinsulin and processing the same to form active, mature insulin and methods of culturing such cells
DK0702085T4 (en) 1994-07-18 2010-04-06 Conzelmann Karl Klaus Prof Dr Recombinant infectious non-segmented negative-stranded RNA virus
FR2723104B1 (en) * 1994-07-27 1996-09-13 Rhone Poulenc Rorer Sa NOVEL TOPOISOMERASE IV, CORRESPONDING NUCLEOTIC SEQUENCES AND USES THEREOF
US5824536A (en) * 1994-08-23 1998-10-20 St. Jude Children's Research Hospital Influenza virus replicated in mammalian cell culture and vaccine production
US5789229A (en) * 1994-09-30 1998-08-04 Uab Research Foundation Stranded RNA virus particles
US5753489A (en) * 1994-11-10 1998-05-19 Immuno Ag Method for producing viruses and vaccines in serum-free culture
JPH11509081A (en) 1994-11-16 1999-08-17 セント ジュード チルドレンズ リサーチ ホスピタル New replication process
US7153510B1 (en) 1995-05-04 2006-12-26 Yale University Recombinant vesiculoviruses and their uses
US5690937A (en) * 1995-06-05 1997-11-25 Aviron Temperature sensitive clustered changed-to-alanine mutants of influenza virus PB2 gene
DE69510207T3 (en) 1995-08-09 2007-02-15 Schweiz. Serum- & Impfinstitut Bern Process for producing infectious minus-stranded RNA viruses
JPH11512609A (en) 1995-09-27 1999-11-02 アメリカ合衆国 Production of infectious respiratory syncytial virus from cloned nucleotide sequence
AU3799797A (en) 1996-07-15 1998-02-09 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Production of attenuated respiratory syncytial virus vaccines from cloned nucleotide sequences
BR9712138A (en) 1996-09-27 2000-01-18 American Cyanamid Co Isolated RNA virus, vaccine, process to immunize an individual to induce protection against a non-segmented, negative, single-stranded RNA virus of the mononegaviral order and to produce RNA virus, isolated nucleic acid molecule and composition.
CA2291216A1 (en) 1997-05-23 1998-11-26 Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services National Institutes Of Health Production of attenuated parainfluenza virus vaccines from cloned nucleotide sequences
WO1999002657A1 (en) 1997-07-11 1999-01-21 Yale University Rhabdoviruses with reengineered coats
US6946291B2 (en) * 1998-04-24 2005-09-20 University Hospitals Of Cleveland Mixed cell diagnostic systems
US6146642A (en) * 1998-09-14 2000-11-14 Mount Sinai School Of Medicine, Of The City University Of New York Recombinant new castle disease virus RNA expression systems and vaccines
US6544785B1 (en) * 1998-09-14 2003-04-08 Mount Sinai School Of Medicine Of New York University Helper-free rescue of recombinant negative strand RNA viruses
DE60033284T3 (en) 1999-07-14 2014-10-30 Icahn School Of Medicine At Mount Sinai IN VITRO RECONSTITUTION OF SEGMENTED NEGATIVE RADIATION RNA VIRUSES
KR100848719B1 (en) 2000-04-28 2008-07-25 세인트 쥬드 칠드런즈 리써치 호스피탈 Dna transfection system for the generation of infectious influenza virus
WO2002006453A2 (en) * 2000-07-18 2002-01-24 Bayer Aktiengesellschaft Regulation of human desc1-like serine protease
US20040241139A1 (en) * 2000-07-20 2004-12-02 Gerd Hobom Recombinant influenza viruses with bicistronic vRNAs coding for two genes in tandem arrangement
ES2806412T3 (en) * 2002-02-13 2021-02-17 Wisconsin Alumni Res Found Signal for packaging of influenza virus vectors
EP1485488A4 (en) * 2002-02-21 2005-03-02 Sinai School Medicine Recombinant negative strand virus rna expression systems and vaccines
US7465456B2 (en) * 2002-04-26 2008-12-16 Medimmune, Llc Multi plasmid system for the production of influenza virus
EP1499348B1 (en) 2002-04-26 2014-10-01 MedImmune, LLC Method for producing infectious influenza b virus in cell culture
US7132523B2 (en) * 2002-07-03 2006-11-07 Ortho-Mcneil Pharmaceutical, Inc. Human PRSS11-Like S2 serine protease and uses thereof
MX282672B (en) * 2002-10-10 2011-01-10 Diversa Corp Proteases, nucleic acids encoding them and methods for making and using them.
WO2005014862A1 (en) * 2003-02-25 2005-02-17 Medimmune Vaccines, Inc. Methods of producing inflenza vaccine compositions
CA2421086A1 (en) * 2003-02-28 2004-08-28 Institut Pasteur Chicken rna polymerase i promoter and its use
US7723094B2 (en) * 2003-05-28 2010-05-25 Wisconsin Alumni Research Foundation Recombinant influenza vectors with a polII promoter and ribozymes for vaccines and gene therapy
AU2004249133B2 (en) 2003-05-28 2009-05-28 Wisconsin Alumni Research Foundation High titer recombinant influenza viruses for vaccines and gene therapy
ATE469972T1 (en) * 2003-12-23 2010-06-15 Medimmune Llc MULTIPLASMID SYSTEM FOR GENERATING THE FLU VIRUS
RU2435855C2 (en) * 2004-12-24 2011-12-10 Солвей Байолоджикалз Б.В. Method for producing replicative influenza virus particles, cell composition (versions), cell culture composition and application thereof
US7790434B2 (en) * 2005-06-21 2010-09-07 Medimmune, Llc Methods and compositions for expressing negative-sense viral RNA in canine cells
EP1917352A4 (en) 2005-06-21 2009-03-25 Medimmune Vaccines Inc Methods and compositions for expressing a heterologous protease
DK2016194T3 (en) * 2006-04-19 2015-06-15 Medimmune Llc METHODS AND COMPOSITIONS FOR EXPRESSION OF VIRAL NEGATIVE SENSE RNA IN DOG CELLS

Also Published As

Publication number Publication date
BRPI0612268A2 (en) 2009-01-27
KR20080024212A (en) 2008-03-17
CA2613284A1 (en) 2007-01-04
NO20080358L (en) 2008-03-03
US8278433B2 (en) 2012-10-02
EP1917361A2 (en) 2008-05-07
WO2007002007A2 (en) 2007-01-04
US20060286591A1 (en) 2006-12-21
AU2006262381A1 (en) 2007-01-04
IL188202A0 (en) 2008-03-20
WO2007002007A3 (en) 2007-06-21
JP2008546408A (en) 2008-12-25
EP1917361A4 (en) 2009-07-29
EP1917352A2 (en) 2008-05-07
EP1917352A4 (en) 2009-03-25
JP2008543331A (en) 2008-12-04
US20110045586A1 (en) 2011-02-24
US20120329150A1 (en) 2012-12-27
US20100203641A1 (en) 2010-08-12
KR20080025171A (en) 2008-03-19
WO2007002008A2 (en) 2007-01-04
WO2007002008A3 (en) 2007-12-27
US8742089B2 (en) 2014-06-03
WO2007002007A8 (en) 2007-05-10
RU2008101367A (en) 2009-08-10
AU2006262380A1 (en) 2007-01-04

Similar Documents

Publication Publication Date Title
CA2613283A1 (en) Methods and compositions for expressing a heterologous protease
AU2007240448B8 (en) Methods and compositions for expressing negative-sense viral RNA in canine cells
US8802417B2 (en) Production of viruses, viral isolates and vaccines
US7790434B2 (en) Methods and compositions for expressing negative-sense viral RNA in canine cells
CN101297029A (en) Methods and compositions for expressing a heterologous protease
AU2013204811B2 (en) Methods and compositions for expressing negative-sense viral rna in canine cells
Rawangkhan et al. Comparison of neuraminidase activity of influenza A virus subtype H5N1 and H1N1 using reverse genetics virus
Marzio et al. Production of viruses, viral isolates and vaccines
MX2008000245A (en) Methods and compositionsfor expressing negative-sense viral rna in canine cells

Legal Events

Date Code Title Description
FZDE Discontinued